1
|
Svobodova B, Moravcova Z, Misiachna A, Novakova G, Marek A, Finger V, Odvarkova J, Pejchal J, Karasova JZ, Netolicky J, Ladislav M, Hrabinova M, Sorf A, Muckova L, Fikejzlova L, Benkova M, Novak M, Prchal L, Capek J, Handl J, Rousar T, Greber KE, Ciura K, Horak M, Soukup O, Korabecny J. Novel tacrine-based multi-target directed Ligands: Enhancing cholinesterase inhibition, NMDA receptor antagonism, and CNS bioavailability for Alzheimer's disease treatment. Eur J Med Chem 2025; 292:117678. [PMID: 40288120 DOI: 10.1016/j.ejmech.2025.117678] [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] [Revised: 04/08/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Alzheimer's disease (AD) is a multifaceted neurodegenerative disorder for which current treatments provide only symptomatic relief, primarily through cholinesterase (ChE) inhibition and N-methyl-d-aspartate receptor (NMDAR) antagonism. To improve therapeutic efficacy and safety, we designed and synthesized 16 novel tacrine derivatives modified at position 7 with various (hetero)aryl groups or deuterium substitution. Initially, in silico screening predicted favorable CNS permeability and oral bioavailability. Subsequent in vitro evaluations demonstrated significant inhibitory potency against acetylcholinesterase (AChE) and butyrylcholinesterase (BChE), with derivatives 5i and 5m displaying particularly promising profiles. Metabolic stability assessed using human liver microsomes revealed enhanced stability for compound 5e, whereas 5i and 5m underwent rapid metabolism. Notably, compound 7 showed improved metabolic stability attributed to deuterium incorporation. The newly synthesized compounds were further tested for antagonistic activity on the GluN1/GluN2B subtype of NMDAR, with compound 5m exhibiting the most potent and voltage-independent inhibition. The ability of these compounds to permeate the blood-brain barrier (BBB) was confirmed through in vitro PAMPA assays. In preliminary hepatotoxicity screening (HepG2 cells), most derivatives exhibited higher cytotoxicity than tacrine, emphasizing the ongoing challenge in hepatotoxicity management. Based on its overall favorable profile, compound 5m advanced to in vivo pharmacokinetic studies in mice, demonstrating efficient CNS penetration, with brain concentrations exceeding plasma levels (brain-to-plasma ratio 2.36), indicating active transport across the BBB. These findings highlight compound 5m as a promising tacrine-based multi-target-directed ligand, supporting further preclinical development as a potential therapeutic candidate for AD.
Collapse
Affiliation(s)
- Barbora Svobodova
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Zuzana Moravcova
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Anna Misiachna
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic; Department of Physiology, Faculty of Science, Charles University in Prague, Albertov 6, Prague 2, 12843, Czech Republic
| | - Gabriela Novakova
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, Prague 6, 166 10, Czech Republic
| | - Ales Marek
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, Prague 6, 166 10, Czech Republic
| | - Vladimir Finger
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic
| | - Jitka Odvarkova
- Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Jaroslav Pejchal
- Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Jana Zdarova Karasova
- Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Jakub Netolicky
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic; Department of Physiology, Faculty of Science, Charles University in Prague, Albertov 6, Prague 2, 12843, Czech Republic
| | - Marek Ladislav
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic; Department of Physiology, Faculty of Science, Charles University in Prague, Albertov 6, Prague 2, 12843, Czech Republic
| | - Martina Hrabinova
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Ales Sorf
- Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic; Department of Social and Clinical Pharmacy, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Lubica Muckova
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Lenka Fikejzlova
- Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Marketa Benkova
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic
| | - Martin Novak
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic
| | - Lukas Prchal
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic
| | - Jan Capek
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentska 573, 532 10, Pardubice, Czech Republic
| | - Jiri Handl
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentska 573, 532 10, Pardubice, Czech Republic
| | - Tomas Rousar
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentska 573, 532 10, Pardubice, Czech Republic
| | - Katarzyna Ewa Greber
- Department of Physical Chemistry, Faculty of Pharmacy, Medical University of Gdansk, Aleja Generała Jozefa Hallera 107, 80-416, Gdansk, Poland
| | - Krzesimir Ciura
- Department of Physical Chemistry, Faculty of Pharmacy, Medical University of Gdansk, Aleja Generała Jozefa Hallera 107, 80-416, Gdansk, Poland; Laboratory of Environmental Chemoinformatics, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308, Gdansk, Poland
| | - Martin Horak
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic
| | - Ondrej Soukup
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic.
| | - Jan Korabecny
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic.
| |
Collapse
|
2
|
Wang M, Zhang X, Zhong L, Zeng L, Li L, Yao P. Understanding autism: Causes, diagnosis, and advancing therapies. Brain Res Bull 2025; 227:111411. [PMID: 40449388 DOI: 10.1016/j.brainresbull.2025.111411] [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: 03/19/2025] [Revised: 05/28/2025] [Accepted: 05/28/2025] [Indexed: 06/03/2025]
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental condition marked by difficulties in social communication, languages, and repetitive behaviors. Its rising prevalence has made it a critical global public health issue. ASD is believed to arise from a combination of genetic and environmental influences. While some gene mutations associated with ASD have been identified, most cases lack clear genetic explanations. Evidence increasingly points to early-life environmental factors as key contributors to ASD, including advanced parental age, maternal diabetes during pregnancy, infections, hormonal imbalances, certain medications, and exposure to air pollution. Currently, ASD diagnosis relies on behavioral assessments, but the absence of specific molecular biomarkers poses significant obstacles to early detection and targeted therapies. Encouragingly, research has identified potential biomarkers, such as neuroimaging classifiers, electroencephalography patterns, eye-tracking data, digital analytics, gene expression profiles, inflammatory and chemokine markers, proteomic and metabolomic profiles, and gut microbiota characteristics. Potential therapeutical strategies under investigation include digital therapies, non-invasive brain stimulation, antioxidants, oxytocin, AVPR1a antagonists, PPAR agonists, and mTOR inhibitors. This review explores ASD across five areas: epidemiological trends, genetic mechanisms, early-life environmental factors and their potential roles, diagnostic biomarkers, and therapeutic approaches.
Collapse
Affiliation(s)
- Min Wang
- Hainan Women and Children's Medical Center, Hainan Medical University, Haikou 570206, PR China
| | - Xiaozhuang Zhang
- Hainan Women and Children's Medical Center, Hainan Medical University, Haikou 570206, PR China
| | - Liyan Zhong
- Hainan Women and Children's Medical Center, Hainan Medical University, Haikou 570206, PR China
| | - Liqin Zeng
- Department of gynecology, Sun Yat-Sen University Affiliated No.8 Hospital, Shenzhen 518033, PR China
| | - Ling Li
- Hainan Women and Children's Medical Center, Hainan Medical University, Haikou 570206, PR China.
| | - Paul Yao
- Hainan Women and Children's Medical Center, Hainan Medical University, Haikou 570206, PR China.
| |
Collapse
|
3
|
Kumar R, Kumari P, Kumar R. Central Nervous System Response Against Ionizing Radiation Exposure: Cellular, Biochemical, and Molecular Perspectives. Mol Neurobiol 2025; 62:7268-7295. [PMID: 39875779 DOI: 10.1007/s12035-025-04712-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: 07/11/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025]
Abstract
Gamma radiation is known to induce several detrimental effects on the nervous system. The hippocampus region, specifically the dentate gyrus (DG) and subventricular zone (SVZ), have been identified as a radiation-sensitive neurogenic niche. Radiation alters the endogenous redox status of neural stem cells (NSCs) and other proliferative cells, especially in the hippocampus region, leading to oxidative stress, neuroinflammation, and cell death. Planned (i.e., radiotherapy of brain tumor patients) or unplanned radiation exposure (i.e., accidental radiation exposure) can induce nonspecific damage to neuronal tissues, resulting in chronic or acute radiation syndrome. Although anatomical alterations in the neuronal tissues have been reported at higher doses of gamma radiation, biochemical and molecular perturbations may be evident even at much lower radiation doses. They may manifest in the form of neuronal deficits and cognitive impairment. In the present review, several molecular events and signaling pathways, such as oxidative stress, neuroinflammation, apoptosis, cognition, neuroplasticity, and neurotoxicity induced in neuronal cells upon ionizing radiation exposure, are reviewed. Furthermore, brain-specific radioprotectors and mitigators that protect normal neuronal cells and tissues against ionizing radiation during radiotherapy of cancer patients or nuclear emergencies are also discussed.
Collapse
Affiliation(s)
- Ravi Kumar
- Radiation Biotechnology Department, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Brig. S.K. Mazumdar Road, Timarpur, Delhi, 110054, India
| | - Pratibha Kumari
- Radiation Biotechnology Department, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Brig. S.K. Mazumdar Road, Timarpur, Delhi, 110054, India
| | - Raj Kumar
- Radiation Biotechnology Department, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Brig. S.K. Mazumdar Road, Timarpur, Delhi, 110054, India.
| |
Collapse
|
4
|
Dhureja M, Munshi A, Kumar P. AMPK as a Therapeutic Target: Advancing Epilepsy Management Through Metabolic Modulation. Mol Neurobiol 2025; 62:7820-7834. [PMID: 39937419 DOI: 10.1007/s12035-025-04745-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/04/2025] [Indexed: 02/13/2025]
Abstract
Epilepsy is often marked by paroxysmal seizures that disrupt the brain's sensory, motor, and psychosocial functions. The underlying pathology is generally believed to involve an imbalance between excitatory and inhibitory neurotransmission. However, a less explored but significant contributor to epilepsy is the collapse of the brain's metabolic and bioenergetic systems. The breakdown of the brain's bioenergetic system leads to the activation of various detrimental downstream signaling cascades that ultimately result in oxidative stress, neuroinflammation, and reduced autophagic flux, all of which impair neuronal-glial communication and precipitate epileptic attacks. This highlights the pressing need for a therapeutic agent to address these complex challenges. Researchers have identified adenosine monophosphate kinase (AMPK) as a potential solution. AMPK acts as the body's primary stress sensor, activated in response to the deficiency of growth factors and nutrient starvation to restore energy homeostasis. AMPK activation also maintains the intricate communication between neurons and glial cells, preserving synaptic plasticity integrity, mitigating mitochondrial damage, and dampening inflammatory signaling cascades. Despite demonstrating significant efficacy in managing a range of peripheral and neurological disorders, the role of AMPK in neurotransmission and epilepsy remains unexplored. This review explores the multifaceted molecular roles of AMPK beyond its traditional metabolic regulatory functions, suggesting that targeting AMPK could provide a novel avenue for drug development in epilepsy treatment.
Collapse
Affiliation(s)
- Maanvi Dhureja
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Bathinda, India.
| |
Collapse
|
5
|
Zaky HS, El-Said NT, Aboutaleb AS, Allam A, Mansour M, Ahmed HI, Abdel-Sattar SA. Mito-TEMPO Mitigates Fibromyalgia Induced by Reserpine in Rats: Orchestration Between SIRT1, Mitochondrial Dynamics, Endoplasmic Reticulum and miRNA-320. Neurochem Res 2025; 50:172. [PMID: 40434586 PMCID: PMC12119751 DOI: 10.1007/s11064-025-04424-9] [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: 02/27/2025] [Revised: 04/28/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025]
Abstract
Fibromyalgia (FM) is a chronic disorder that lacks both well-defined underlying causes and effective treatments. Mito-TEMPO (MIT) is a mitochondrial-specific antioxidant that has demonstrated benefits in many cancerous, renal, cardiovascular, and neurodegenerative disorders. However, the therapeutic effect of MIT on FM remains ambiguous. The objective of the current work is to illuminate the use of MIT for FM and its prospective mechanisms. Here, we used the FM rat model induced by three days of subcutaneous reserpine injection (1 mg/kg) and examined the role of MIT on SIRT1 activation and other implicated molecular pathways. Behavioral tests showed that MIT (0.7 mg/kg) can effectively alleviate the locomotor, nociceptive, and depressive-like behaviors in reserpinized rats, an effect that simultaneously reconciles the balance of monoamines in the rat brain. Western blot analysis showed that MIT up-regulates SIRT1 and improves the expression of mitochondrial dynamics proteins (DRP1 and OPA1) and the endoplasmic reticulum protein (CHOP). Furthermore, MIT treatment significantly enhanced the SOD and CAT activities and decreased the brain contents of NF-κB, TNF-α, and BAX, but significantly enriching the Bcl-2 content. Lastly, MIT treatment significantly reduced the genetic expression of miRNA-320 following RES treatment. All the measured parameters showed a significant correlation with SIRT1 expression. Our results suggest that MIT provides antioxidant, anti-apoptotic, and anti-inflammatory impacts on the FM rat model, with proposed mechanisms involved activating the SIRT1 pathway to regulate mitochondrial dynamics, endoplasmic reticulum stress, as well as miRNA-320. Thus, MIT has the potential to be an effectual drug candidate for FM treatment.
Collapse
Affiliation(s)
- Heba S Zaky
- Pharmacology and Toxicology Department, Faculty of Pharmacy for Girls, Al-Azhar University, Nasr City, Cairo, P.N.11754, Egypt
| | - Nermin T El-Said
- Pharmacology and Toxicology Department, Faculty of Pharmacy for Girls, Al-Azhar University, Nasr City, Cairo, P.N.11754, Egypt
| | - Amany S Aboutaleb
- Pharmacology and Toxicology Department, Faculty of Pharmacy for Girls, Al-Azhar University, Nasr City, Cairo, P.N.11754, Egypt
| | - Albatoul Allam
- Pharmacology and Toxicology Department, Faculty of Pharmacy for Girls, Al-Azhar University, Nasr City, Cairo, P.N.11754, Egypt.
| | - Mona Mansour
- Pharmacology and Toxicology Department, Faculty of Pharmacy for Girls, Al-Azhar University, Nasr City, Cairo, P.N.11754, Egypt
| | - Hebatalla I Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy for Girls, Al-Azhar University, Nasr City, Cairo, P.N.11754, Egypt
| | - Somaia A Abdel-Sattar
- Pharmacology and Toxicology Department, Faculty of Pharmacy for Girls, Al-Azhar University, Nasr City, Cairo, P.N.11754, Egypt
| |
Collapse
|
6
|
Kazemi M, Esmaeili-Mahani S, Abbasnejad M, Sheibani V. Neurotrophic factor neuritin ameliorates streptozotocin-induced Alzheimer's disease-like impairment of memory, neuroinflammation, apoptotic factors and compensates hippocampal neuritin expression. Behav Brain Res 2025; 486:115542. [PMID: 40127821 DOI: 10.1016/j.bbr.2025.115542] [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: 08/24/2024] [Revised: 03/13/2025] [Accepted: 03/13/2025] [Indexed: 03/26/2025]
Abstract
Alzheimer's disease (AD) is the main cause of dementia in the elderly, and is becoming one of the most expensive and deadly diseases. Deficiency of neurotrophic factors signaling is an important cause of this disease. Therefore, we investigated whether neuritin as a neurotrophic factor can have a neuroprotective effect against streptozotocin (STZ)-induced rat model of AD. The animals were bilaterally injected with intra hippocampal-STZ (3 mg/kg). Different concentrations of neuritin (0.5, 1, 1.5 µg/rat) were administrated 15 min before STZ injection. After 14 days, the rats were evaluated for cognitive performance using novel object recognition (NOR), open field and Morris water maze (MWM) tests and then sacrificed for biochemical analysis (by real-time PCR and western blot examinations). The results demonstrated that the STZ- induced learning and memory impairments were significantly prevented by 1.5 µg neuritin. Moreover, the increased levels of inflammatory factors (NF-κb, TNF-α and IL-1β) and apoptotic parameters (cytochrome c and caspase‑3) in STZ- treated rats were also significantly decreased by neuritin. In addition, hippocampal neuritin gene expression was downregulated by STZ injection, which was reversed by intra hippocampal neuritin injection. In conclusion, the present study suggests that neuritin prevents cognitive defects in AD rat model and its expression level is associated with cognitive resilience.
Collapse
Affiliation(s)
- Mandana Kazemi
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Saeed Esmaeili-Mahani
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran; Kerman Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mehdi Abbasnejad
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Vahid Sheibani
- Kerman Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
7
|
Ghosh S, Das Sarma J. The age-dependent neuroglial interaction with peripheral immune cells in coronavirus-induced neuroinflammation with a special emphasis on COVID-19. Biogerontology 2025; 26:111. [PMID: 40380990 DOI: 10.1007/s10522-025-10252-9] [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: 02/28/2025] [Accepted: 05/02/2025] [Indexed: 05/19/2025]
Abstract
Neurodegenerative diseases are chronic progressive disorders that impair memory, cognition, and motor functions, leading to conditions such as dementia, muscle weakness, and speech difficulties. Aging disrupts the stringent balance between pro- and anti-inflammatory cytokines, increasing neuroinflammation, which contributes to neurodegenerative diseases. The aging brain is particularly vulnerable to infections due to a weakened and compromised immune response and impaired integrity of the blood-brain barrier, allowing pathogens like viruses to trigger neurodegeneration. Coronaviruses have been linked to both acute and long-term neurological complications, including cognitive impairments, psychiatric disorders, and neuroinflammation. The virus can induce a cytokine storm, damaging the central nervous system (CNS) and worsening existing neurological conditions. Though its exact mechanism of neuroinvasion remains elusive, evidence suggests it disrupts the blood-brain barrier and triggers immune dysregulation, leading to persistent neurological sequelae in elderly individuals. This review aims to understand the interaction between the peripheral immune system and CNS glial cells in aged individuals, which is imperative in addressing coronavirus-induced neuroinflammation and concomitant neurodegeneration.
Collapse
Affiliation(s)
- Satavisha Ghosh
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohanpur, Kolkata, 741246, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohanpur, Kolkata, 741246, India.
- Department of Ophthalmology, University of Pennsylvania, 19104, Philadelphia, PA, USA.
| |
Collapse
|
8
|
Qi Y, Xie S, Chen J, Zhang C, Ma X, Yu Y, Yu X, Wang Y. Gut microbiota regulation by Lactiplantibacillus plantarum SG5 enhances mitochondrial function in Parkinson's disease mice via the GLP-1/PGC-1α pathway. J Nutr Biochem 2025:109954. [PMID: 40368220 DOI: 10.1016/j.jnutbio.2025.109954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/24/2025] [Accepted: 05/05/2025] [Indexed: 05/16/2025]
Abstract
Motor dysfunction constitutes a prominent characteristic of Parkinson's disease (PD), a neurodegenerative disorder associated with compromised mitochondrial activity, perturbed gut microbial composition, and neuronal loss. In this study, we examined the regulatory mechanisms of Lactiplantibacillus plantarum SG5 (SG5) on mitochondrial function in PD mouse models, with a particular emphasis on its interaction with the GLP-1/PGC-1α pathway. Findings revealed that MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, MPTP) induced (male 6-8 weeks C57BL/6 mice) motor impairments and damage to dopaminergic (DA) neurons in PD mice, resulting in mitochondrial dysfunction, decreased mitochondrial biogenesis, disrupted dynamics, and autophagy, while promoting fission and apoptosis. Additionally, MPTP modified gut microbial diversity and community structure. Nevertheless, supplementation with SG5 alleviated motor deficits and DA neurons damage in PD mice, enhancing mitochondrial quality by elevating PGC-1α expression and restoring biogenesis, dynamics, and autophagy levels. Mechanistic investigations demonstrated that SG5 increased colonic GLP-1 expression, suggesting that GLP-1 might regulate mitochondrial function via the GLP-1R-mediated PGC-1α. Furthermore, SG5 counteracted MPTP-induced gut dysbiosis. Notably, both GLP-1R antagonists and PGC-1α inhibitors attenuated the protective effects of SG5 in PD mice. In conclusion, L. plantarum SG5 may enhance mitochondrial function in the substantia nigra (SN) of PD mice through the GLP-1/PGC-1α pathway, potentially delaying neurodegeneration. Its mechanism is closely related to the regulation of the gut microenvironment and GLP-1 levels, presenting novel microbiota-based therapeutic targets for PD.
Collapse
Affiliation(s)
- Yueyan Qi
- Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Hebei Key Laboratory of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Siyou Xie
- Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Hebei Key Laboratory of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Jinhu Chen
- Department of endocrinology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Cancan Zhang
- Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Hebei Key Laboratory of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Xin Ma
- Thankcome Biotechnology (Su Zhou) Co., Suzhou, China
| | - Yang Yu
- Thankcome Biotechnology (Su Zhou) Co., Suzhou, China
| | - Xueping Yu
- Thankcome Biotechnology (Su Zhou) Co., Suzhou, China
| | - Yanqin Wang
- Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Hebei Key Laboratory of Physiology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China.
| |
Collapse
|
9
|
Silla A, Punzo A, Caliceti C, Barbalace MC, Hrelia S, Malaguti M. The Role of Antioxidant Compounds from Citrus Waste in Modulating Neuroinflammation: A Sustainable Solution. Antioxidants (Basel) 2025; 14:581. [PMID: 40427463 PMCID: PMC12108332 DOI: 10.3390/antiox14050581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2025] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
In normal conditions, neuroinflammation induces microglia and astrocyte activation to maintain brain homeostasis. However, excessive or prolonged neuroinflammation can inflict harmful damage on brain tissue. Numerous factors can trigger chronic neuroinflammation, ultimately leading to neurodegeneration. In this context, considering the pressing need for novel, natural approaches to mitigate neuroinflammatory damage, attention has turned to unconventional sources such as agricultural by-products. Citrus fruits are widely consumed globally, producing substantial waste, including peels, seeds, and pulp. Traditionally regarded as agricultural waste, these by-products are now recognized as valuable reservoirs of bioactive compounds, including flavonoids, carotenoids, terpenoids, and limonoids. Among these, citrus polyphenols-particularly flavanones like hesperidin, naringenin, and eriocitrin-have emerged as potent modulators of neuroinflammatory pathways through their multifaceted interactions with cellular antioxidant systems, pro-inflammatory signaling cascades, neurovascular integrity, and gut-brain axis dynamics. This review aims to characterize the key molecules present in citrus waste and synthesizes preclinical and clinical evidence to elucidate the biochemical mechanisms underlying neuroinflammation in neurodegenerative disorders.
Collapse
Affiliation(s)
- Alessia Silla
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy; (A.S.); (A.P.); (C.C.)
| | - Angela Punzo
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy; (A.S.); (A.P.); (C.C.)
| | - Cristiana Caliceti
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy; (A.S.); (A.P.); (C.C.)
| | - Maria Cristina Barbalace
- Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, 47921 Rimini, Italy; (M.C.B.); (M.M.)
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, 47921 Rimini, Italy; (M.C.B.); (M.M.)
| | - Marco Malaguti
- Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, 47921 Rimini, Italy; (M.C.B.); (M.M.)
| |
Collapse
|
10
|
Fang YW, Lin HC, Wang C, Lin CY. Glyphosate Exposure, Oxidative Stress, Mitochondrial Dysfunction, and Mortality Risk in US Adults: Insights from the National Health and Nutrition Examination Survey. TOXICS 2025; 13:373. [PMID: 40423452 DOI: 10.3390/toxics13050373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2025] [Revised: 05/01/2025] [Accepted: 05/02/2025] [Indexed: 05/28/2025]
Abstract
Purpose: Glyphosate and glyphosate-based herbicides (GBHs) are widely used across the globe. Experimental research indicates that these herbicides may elevate oxidative stress and impair mitochondrial function. However, the relationship between glyphosate exposure, oxidative stress, and mitochondrial function remains poorly characterized in epidemiological studies. In particular, the role of oxidative stress and mitochondrial function biomarkers in mediating the mortality risk associated with glyphosate exposure in nationally representative populations is not well understood. Approach and Results: In this study, we utilized data from the 2013-2014 National Health and Nutrition Examination Survey (NHANES), encompassing 1464 participants aged 18 years and older. This dataset was linked to mortality records from the National Center for Health Statistics (NCHS), with follow-up data extending through 2019. The primary objective was to examine the associations between urinary glyphosate levels and biomarkers of oxidative stress and mitochondrial function-specifically pyrazino-s-triazine derivative of 4-α-hydroxy-5-methyl-tetrahydrofolate (MeFox) and methylmalonic acid (MMA)-and to evaluate the role of these biomarkers in influencing glyphosate-related mortality outcomes. Results: Urinary glyphosate levels were positively associated with serum MMA and MeFox in weighted multiple linear regression models. For MMA, glyphosate showed significant positive associations in both adjusted models (Model 2: β = 0.061, p = 0.001). Similarly, urinary glyphosate was strongly associated with MeFox in all models (Model 2: β = 0.215, p < 0.001). During a median follow-up of 69.57 months, 116 deaths occurred, including 44 from cardiovascular causes. Glyphosate was not significantly associated with all-cause or cardiovascular mortality in the overall population. However, subgroup analysis revealed significant associations in individuals with higher MeFox levels (≥50th percentile) for all-cause mortality (HR = 1.395, p = 0.027) and borderline associations for cardiovascular mortality (HR = 1.367, p = 0.051). When adjusted for MMA, glyphosate was significantly associated with increased all-cause mortality in participants with MMA levels below the 50th percentile (HR = 2.679, p = 0.001), with a significant interaction between glyphosate and MMA for all-cause (p = 0.002) and cardiovascular mortality (p = 0.038). Conclusions: In this comprehensive analysis of NHANES data, urinary glyphosate levels were associated with biomarkers of oxidative stress and mitochondrial function. While no overall mortality associations were observed, glyphosate exposure was linked to increased all-cause mortality in subgroups with lower MMA or higher MeFox levels. These findings highlight the role of oxidative stress and mitochondrial function in glyphosate-related health risks and the need for further research to identify vulnerable populations.
Collapse
Affiliation(s)
- Yu-Wei Fang
- Division of Nephrology, Department of Internal Medicine, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Hsuan-Cheng Lin
- School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Chikang Wang
- Department of Environmental Engineering and Health, Yuanpei University of Medical Technology, Hsinchu 300, Taiwan
| | - Chien-Yu Lin
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
- Department of Environmental Engineering and Health, Yuanpei University of Medical Technology, Hsinchu 300, Taiwan
- Department of Internal Medicine, En Chu Kong Hospital, Taipei 237, Taiwan
| |
Collapse
|
11
|
Naeimi H, Taheri M, Ghafouri H, Mohammadi A. Investigation of Thiazolidine-2,4-Dione Derivatives as Acetylcholinesterase Inhibitors: Synthesis, In Vitro Biological Activities and In Silico Studies. ChemistryOpen 2025; 14:e202400294. [PMID: 39797425 DOI: 10.1002/open.202400294] [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: 08/09/2024] [Revised: 12/04/2024] [Indexed: 01/13/2025] Open
Abstract
The inhibition of acetylcholinesterase (AChE), an enzyme responsible for the inactivation and decrease in acetylcholine in the cholinergic pathway, has been considered an attractive target for small-molecule drug discovery in Alzheimer's disease (AD) therapy. In the present study, a series of TZD derivatives were designed, synthesized, and studied for drug likeness, blood-brain barrier (BBB) permeability, and adsorption, distribution, metabolism, excretion, and toxicity (ADMET). Additionally, docking studies of the designed compounds were performed on AChE. Additionally, all the TZD derivatives (CHT1-5) showed an acceptable affinity for AChE inhibition, and the results showed convincing binding modes in the active site of AChE. Among them, 5-(4-methoxybenzylidene) thiazolidine-2,4-dione (CHT1) was identified as the most potent AChE inhibitor (IC50 of 165.93 nM) with the highest antioxidant activity. Following the exposure of PC12 cells to Aβ1-42 (100 μM), a marked reduction in cell survival was observed. Pretreatment of PC12 cells with TZD derivatives had a neuroprotective effect and significantly enhanced cell survival in response to Aβ-induced toxicity. Western blotting analysis revealed that CHT1 (5 and 8 μM) downregulated p-Tau and HSP70 expression levels. The results indicate that CHT1 is a promising and effective AchE-I that could be utilized as a powerful candidate against AD.
Collapse
Affiliation(s)
- Hanane Naeimi
- Department of Biology, Faculty of Basic Sciences, University of Guilan, Rasht, 4193833697, Iran
| | - Maryam Taheri
- Department of Biology, Faculty of Basic Sciences, University of Guilan, Rasht, 4193833697, Iran
| | - Hossein Ghafouri
- Department of Biology, Faculty of Basic Sciences, University of Guilan, Rasht, 4193833697, Iran
| | - Asadollah Mohammadi
- Department of Chemistry, Faculty of Sciences, University of Guilan, Rasht, 4193833697, Iran
| |
Collapse
|
12
|
D'Alessandro MCB, Kanaan S, Geller M, Praticò D, Daher JPL. Mitochondrial dysfunction in Alzheimer's disease. Ageing Res Rev 2025; 107:102713. [PMID: 40023293 DOI: 10.1016/j.arr.2025.102713] [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: 11/06/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease characterized by progressive cognitive decline and distinct neuropathological features. The absence of a definitive cure presents a significant challenge in neurology and neuroscience. Early clinical manifestations, such as memory retrieval deficits and apathy, underscore the need for a deeper understanding of the disease's underlying mechanisms. While amyloid-β plaques and tau neurofibrillary tangles have dominated research efforts, accumulating evidence highlights mitochondrial dysfunction as a central factor in AD pathogenesis. Mitochondria, essential cellular organelles responsible for energy production necessary for neuronal function become impaired in AD, triggering several cellular consequences. Factors such as oxidative stress, disturbances in energy metabolism, failures in the mitochondrial quality control system, and dysregulation of calcium release are associated with mitochondrial dysfunction. These abnormalities are closely linked to the neurodegenerative processes driving AD development and progression. This review explores the intricate relationship between mitochondrial dysfunction and AD pathogenesis, emphasizing its role in disease onset and progression, while also considering its potential as a biomarker and a therapeutic target.
Collapse
Affiliation(s)
- Maria Clara Bila D'Alessandro
- Universidade Federal Fluminense, Faculty of Medicine, Desembargador Athayde Parreiras road 100, Niterói, Rio de Janeiro, Brazil.
| | - Salim Kanaan
- Universidade Federal Fluminense, Faculty of Medicine, Department of Pathology, Marquês do Paraná road, 303, 2nd floor, Niterói, Rio de Janeiro, Brazil.
| | - Mauro Geller
- Unifeso, Department of Immunology and Microbiology, Alberto Torres avenue 111, Teresópolis, Rio de Janeiro, Brazil
| | - Domenico Praticò
- Department of Neurosciences, Lewis Katz School of Medicine. Temple University, 3500 North Broad Street, Philadelphia, PA, United States.
| | - João Paulo Lima Daher
- Universidade Federal Fluminense, Faculty of Medicine, Department of Pathology, Marquês do Paraná road, 303, 2nd floor, Niterói, Rio de Janeiro, Brazil.
| |
Collapse
|
13
|
Xue X, Du J, Hussain SA, Maddu N, Xiong J. Impact of Transchalcone on Neurological Outcomes in Cerebral Ischemia-reperfusion Injury in Rat: Role of AMP-activated Protein Kinase-mitochondrial Signaling Pathways. JOURNAL OF PHYSIOLOGICAL INVESTIGATION 2025; 68:168-175. [PMID: 40223319 DOI: 10.4103/ejpi.ejpi-d-25-00004] [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: 01/16/2024] [Accepted: 02/27/2025] [Indexed: 04/15/2025]
Abstract
ABSTRACT Cerebral ischemia-reperfusion (CIR) injury results in significant secondary brain damage after ischemic stroke due to oxidative stress, mitochondrial dysfunction, and neuroinflammation. Transchalcone (TCH), a polyphenolic compound, exhibits antioxidant and anti-inflammatory properties that may contribute to neuroprotection. The present study investigated the potential protective effects of TCH in a rat model of CIR, focusing on its impact on the activation of AMP-activated protein kinase (AMPK) pathway, mitochondrial function, and inflammatory mediators. Sixty adult Sprague-Dawley rats were randomly divided into five groups of Control, CIR (ischemia-reperfusion only), CIR+TCH (CIR with TCH), CIR+CC (CIR with compound C), and CIR+CC+TCH (CIR with compound C plus TCH). TCH (100 μg/kg b.w per day) was given intraperitoneally over 7 days before CIR injury to animals. Middle cerebral artery occlusion was performed for 60 min to induce cerebral ischemia, and then blood flow was restored (reperfusion) for 24 h. Neuromotor function was assessed using neurological scoring, rotarod, and grid tests. The infarct volumes were determined using 2,3,5-triphenyltetrazolium chloride staining. Mitochondrial function was evaluated using fluorometric and calorimetric methods. Oxidative stress and inflammatory mediators were measured by enzyme-linked immunosorbent assay. Protein expression was analyzed using Western blotting. CIR significantly impaired neuromotor function, increased infarct volume, elevated mitochondrial reactive oxygen species (ROS) levels, and disrupted adenosine triphosphate (ATP) synthesis and manganese superoxide dismutase (Mn-SOD) activity. It also heightened pro-inflammatory cytokines interleukin-1β (IL-1β), tumor necrosis factor-alpha, and nuclear factor kappa B levels while reducing the anti-inflammatory IL-10 level. TCH treatment significantly attenuated CIR outcomes by promoting AMPK phosphorylation, upregulating peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) and nuclear factor erythroid 2-related factor 2 (NRF2) expression, reducing mitochondrial ROS, improving ATP production and Mn-SOD activity, and suppressing pro-inflammatory cytokine mediators while increasing IL-10. Co-treatment with compound C (a selective AMPK inhibitor) significantly diminished the protective effects of TCH, confirming the contribution of AMPK signaling in its neuroprotective mechanism. TCH provides significant neuroprotection against CIR injury by activating AMPK/PGC-1α and AMPK/NRF2 signaling, preserving mitochondrial function, and modulating inflammation. These findings highlight the therapeutic potential of TCH for ischemic stroke management.
Collapse
Affiliation(s)
- Xiuyun Xue
- Department of Neurology, Xi'an Central Hospital, Xi'an, China
| | - Jingjing Du
- Department of Neurology, Xi'an Central Hospital, Xi'an, China
| | - Shaik Althaf Hussain
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Narendra Maddu
- Department of Biochemistry, Sri Krishnadevaraya University, Anantapur, Andhra Pradesh, India
| | - Jing Xiong
- Department of Neurology, Xi'an Central Hospital, Xi'an, China
| |
Collapse
|
14
|
Qu W, Yan G, Du Y, Zhou X, Huang C, Li B, Zhou J, Li Q. Crosstalk Between Mitochondrial DNA and Immune Response: Focus on Autism Spectrum Disorder. Mol Neurobiol 2025; 62:5629-5639. [PMID: 39589631 DOI: 10.1007/s12035-024-04637-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: 07/07/2024] [Accepted: 11/16/2024] [Indexed: 11/27/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by multiple dysfunctions in behavior, the nervous system, and the immune system. Increasing evidence suggests that mitochondrial DNA (mtDNA) plays a crucial role in the pathology of ASD. In clinical practice, altered mtDNA levels have been observed in various tissues of individuals with ASD. Mutation or oxidation of mtDNA is also closely related to the immune response associated with the pathology of autism. With mtDNA identified as a causal factor, much interest has focused on how its production affects neurodevelopment and neurophysiology. Here, we review how mtDNA leads to dysfunction of cellular mitochondria and immune response. We also illustrate the relationship between mtDNA alterations and the pathology of autism. Finally, we discuss the existing evidence on cell-free mtDNA associated with ASD and look forward to its application in clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Wenxuan Qu
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China
| | - Ge Yan
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China
| | - Yajuan Du
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China
| | - Xinyang Zhou
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China
| | - Chutian Huang
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China
| | - Bei Li
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China
| | - Junmei Zhou
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China
| | - Qian Li
- Department of Central Laboratory, School of Medicine, Shanghai Children's Hospital, Shanghai Jiaotong University, 355 Luding Road, Shanghai, 200062, Putuo District, China.
| |
Collapse
|
15
|
Ma Y, Song R, Duan C. Mitochondrial quality control and transfer communication in neurological disorders and neuroinflammation. Front Immunol 2025; 16:1542369. [PMID: 40356918 PMCID: PMC12066325 DOI: 10.3389/fimmu.2025.1542369] [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: 12/09/2024] [Accepted: 04/08/2025] [Indexed: 05/15/2025] Open
Abstract
Mitochondria, as the primary energy factories of cells, play a pivotal role in maintaining nervous system function and regulating inflammatory responses. The balance of mitochondrial quality control is critical for neuronal health, and disruptions in this balance are often implicated in the pathogenesis of various neurological disorders. Mitochondrial dysfunction not only exacerbates energy deficits but also triggers neuroinflammation through the release of damage-associated molecular patterns (DAMPs), such as mitochondrial DNA (mtDNA) and reactive oxygen species (ROS). This review examines the mechanisms and recent advancements in mitochondrial quality control in neurological diseases, focusing on processes such as mitochondrial fusion and fission, mitophagy, biogenesis, and protein expression regulation. It further explores the role of mitochondrial dysfunction and subsequent inflammatory cascades in conditions such as ischemic and hemorrhagic stroke, neurodegenerative diseases and brain tumors. Additionally, emerging research highlights the significance of mitochondrial transfer mechanisms, particularly intercellular transfer between neurons and glial cells, as a potential strategy for mitigating inflammation and promoting cellular repair. This review provides insights into the molecular underpinnings of neuroinflammatory pathologies while underscoring the translational potential of targeting mitochondrial quality control for therapeutic development.
Collapse
Affiliation(s)
| | | | - Chenyang Duan
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
16
|
Giordano L, Ware SA, Lagranha CJ, Kaufman BA. Mitochondrial DNA signals driving immune responses: Why, How, Where? Cell Commun Signal 2025; 23:192. [PMID: 40264103 PMCID: PMC12012978 DOI: 10.1186/s12964-025-02042-0] [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: 10/29/2024] [Accepted: 01/14/2025] [Indexed: 04/24/2025] Open
Abstract
There has been a recent expansion in our understanding of DNA-sensing mechanisms. Mitochondrial dysfunction, oxidative and proteostatic stresses, instability and impaired disposal of nucleoids cause the release of mitochondrial DNA (mtDNA) from the mitochondria in several human diseases, as well as in cell culture and animal models. Mitochondrial DNA mislocalized to the cytosol and/or the extracellular compartments can trigger innate immune and inflammation responses by binding DNA-sensing receptors (DSRs). Here, we define the features that make mtDNA highly immunogenic and the mechanisms of its release from the mitochondria into the cytosol and the extracellular compartments. We describe the major DSRs that bind mtDNA such as cyclic guanosine-monophosphate-adenosine-monophosphate synthase (cGAS), Z-DNA-binding protein 1 (ZBP1), NOD-, LRR-, and PYD- domain-containing protein 3 receptor (NLRP3), absent in melanoma 2 (AIM2) and toll-like receptor 9 (TLR9), and their downstream signaling cascades. We summarize the key findings, novelties, and gaps of mislocalized mtDNA as a driving signal of immune responses in vascular, metabolic, kidney, lung, and neurodegenerative diseases, as well as viral and bacterial infections. Finally, we define common strategies to induce or inhibit mtDNA release and propose challenges to advance the field.
Collapse
Affiliation(s)
- Luca Giordano
- Center for Metabolism and Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus-Liebig-University, Giessen, Germany.
| | - Sarah A Ware
- Center for Metabolism and Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Claudia J Lagranha
- Center for Metabolism and Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brett A Kaufman
- Center for Metabolism and Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
17
|
Volarevic A, Harrell CR, Arsenijevic A, Djonov V, Volarevic V. Therapeutic Potential of Mesenchymal Stem Cell-Derived Extracellular Vesicles in the Treatment of Parkinson's Disease. Cells 2025; 14:600. [PMID: 40277925 PMCID: PMC12025905 DOI: 10.3390/cells14080600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by the gradual loss of dopamine-producing neurons. Oxidative stress, mitochondrial dysfunction, detrimental immune response, and neuroinflammation are mainly responsible for the injury and degeneration of dopaminergic neurons in the brains of patients suffering from PD. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have emerged as a promising therapeutic approach for treating PD due to their ability to suppress the activation of inflammatory immune cells and enhance the viability and function of dopamine-producing neurons. MSC-EVs can easily bypass the blood-brain barrier and deliver their cargo (neuroprotective factors, immunosuppressive proteins, and microRNAs) to injured dopamine-producing neurons and brain-infiltrated inflammatory immune cells. A large number of recently published experimental studies demonstrated that MSC-EVs efficiently alleviated PD-related motor and behavioral deficits in animal models, indicating that MSC-EVs should be considered as potentially new therapeutic agents for the treatment of PD. Accordingly, in this review article, we summarized current knowledge about the therapeutic potential of MSCs-EVs in the treatment of PD, paving the way for their future clinical use in the treatment of neurodegenerative and neuroinflammatory disorders.
Collapse
Affiliation(s)
- Ana Volarevic
- Departments of Psychology, Center for Research on Harmful Effects of Biological and Chemical Hazards, Faculty of Medical Sciences University of Kragujevac, 69 Svetozara Markovica Street, 34000 Kragujevac, Serbia;
| | - Carl Randall Harrell
- Regenerative Processing Plant, LLC, 34176 US Highway 19 N, Palm Harbor, FL 34684, USA
| | - Aleksandar Arsenijevic
- Departments of Genetics, Microbiology and Immunology, Center for Research on Harmful Effects of Biological and Chemical Hazards, Faculty of Medical Sciences University of Kragujevac, 69 Svetozara Markovica Street, 34000 Kragujevac, Serbia;
| | - Valentin Djonov
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland;
| | - Vladislav Volarevic
- Departments of Genetics, Microbiology and Immunology, Center for Research on Harmful Effects of Biological and Chemical Hazards, Faculty of Medical Sciences University of Kragujevac, 69 Svetozara Markovica Street, 34000 Kragujevac, Serbia;
| |
Collapse
|
18
|
Chu CS, Chen YT, Sun WC, Liang WZ. Investigate the protective effects of eicosapentaenoic acid in human astrocytes of oxidative stress damage and explore its underlying mechanisms. Mol Biol Rep 2025; 52:391. [PMID: 40232525 DOI: 10.1007/s11033-025-10481-8] [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: 12/28/2024] [Accepted: 03/27/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND The importance of fatty acids in human health and their potential in treating various brain diseases is increasingly acknowledged. Research indicates that ultra-long-chain fatty acids adversely affect dietary habits, while omega (ω)-3 polyunsaturated fatty acids confer health benefits. Eicosapentaenoic acid (EPA), an ω-3 polyunsaturated fatty acid, manifests diverse protective activities, including anti-oxidative effects and the attenuation of brain diseases. Previous studies have suggested that EPA can alleviate oxidative stress and forestall diseases stemming from oxidative damage. Nevertheless, EPA's precise antioxidant mechanism and signaling pathway in human astrocytes remain elusive. To address this knowledge gap, we established an H2O2-induced oxidative damage model in Gibco® Human Astrocytes (GHA cells) and elucidated the underlying mechanisms and signaling pathways. METHODS AND RESULTS Our assessments included cell viability through the CCK-8 assay, morphological examination via microscopy, ROS quantification using the DCFH-DA fluorescent probe, GSH content evaluation with the CMF-DA fluorescent probe, and protein expression analysis for antioxidant and apoptotic markers through Western blotting. The results showed that pretreatment with 3 µM of EPA countered the cytotoxicity, ROS production, and GSH depletion caused by H2O2 (250 µM) in GHA cells. Additionally, EPA pretreatment effectively reduced the cytotoxicity and oxidative stress resulting from H2O2 by modulating the Nrf2/HO-1/NQO1 and Bax/Bcl-2/caspase-9/caspase-3 signaling pathways in GHA cells. CONCLUSION These findings enhance our understanding of EPA's antioxidant mechanisms in the oxidative stress model of human astrocytes, illuminate the interplay between antioxidant and apoptotic signals, and offer promise for exploring potential preventive and therapeutic interventions for brain diseases.
Collapse
Affiliation(s)
- Che-Sheng Chu
- Department of Psychiatry, Kaohsiung Veterans General Hospital, Kaohsiung, 813414, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan
| | - Ying-Tso Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan
- Department of Neurosurgery, Kaohsiung Veterans General Hospital, Kaohsiung, 813414, Taiwan
| | - Wei-Chih Sun
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, 813414, Taiwan
| | - Wei-Zhe Liang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 813414, Taiwan.
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung County, 90741, Taiwan.
| |
Collapse
|
19
|
Nevoit G, Jarusevicius G, Potyazhenko M, Mintser O, Bumblyte IA, Vainoras A. Mitochondrial Dysfunction and Atherosclerosis: The Problem and the Search for Its Solution. Biomedicines 2025; 13:963. [PMID: 40299559 PMCID: PMC12024619 DOI: 10.3390/biomedicines13040963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/24/2025] [Accepted: 04/09/2025] [Indexed: 05/01/2025] Open
Abstract
Background/Objectives: This review has been prepared to promote interest in the interdisciplinary study of mitochondrial dysfunction (MD) and atherosclerosis. This review aims to describe the state of this problem and indicate the direction for further implementation of this knowledge in clinical medicine. Methods: Extensive research of the literature was implemented to elucidate the role of the molecular mechanisms of MD in the pathogenesis of atherosclerosis. Results: A view on the pathogenesis of atherosclerosis through the prism of knowledge about MD is presented. MD is the cause and primary mechanism of the onset and progression of atherosclerosis. It is proposed that this problem be considered in the context of a continuum. Conclusions: MD and atherosclerosis are united by common molecular mechanisms of pathogenesis. Knowledge of MD should be used to argue for a healthy lifestyle as the primary way to prevent atherosclerosis. The development of new approaches to diagnosing and treating MD in atherosclerosis is an urgent task and challenge for modern science.
Collapse
Affiliation(s)
- Ganna Nevoit
- Laboratory for Automatization of Cardiovascular Investigations, Cardiology Institute, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Gediminas Jarusevicius
- Laboratory for Automatization of Cardiovascular Investigations, Cardiology Institute, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Maksim Potyazhenko
- Department of Internal Medicine and Emergency Medicine, Poltava State Medical University, 36011 Poltava, Ukraine
| | - Ozar Mintser
- Department of Fundamental Disciplines and Informatics, Shupyk National Healthcare University of Ukraine, 04112 Kyiv, Ukraine
| | - Inga Arune Bumblyte
- Department of Nephrology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Alfonsas Vainoras
- Laboratory for Automatization of Cardiovascular Investigations, Cardiology Institute, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| |
Collapse
|
20
|
Kurhaluk N, Kamiński P, Bilski R, Kołodziejska R, Woźniak A, Tkaczenko H. Role of Antioxidants in Modulating the Microbiota-Gut-Brain Axis and Their Impact on Neurodegenerative Diseases. Int J Mol Sci 2025; 26:3658. [PMID: 40332186 PMCID: PMC12027284 DOI: 10.3390/ijms26083658] [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: 03/14/2025] [Revised: 03/27/2025] [Accepted: 04/10/2025] [Indexed: 05/08/2025] Open
Abstract
This narrative review presents the role of antioxidants in regulating the gut microbiota and the impact on the gut-brain axis, with a particular focus on neurodegenerative diseases, such as Alzheimer's (AD) and Parkinson's disease (PD). These diseases are characterised by cognitive decline, motor dysfunction, and neuroinflammation, all of which are significantly exacerbated by oxidative stress. This review elucidates the contribution of oxidative damage to disease progression and explores the potential of antioxidants to mitigate these pathological processes through modulation of the gut microbiota and associated pathways. Based on recent studies retrieved from reputable databases, including PubMed, Web of Science, and Scopus, this article outlines the mechanisms by which antioxidants influence gut health and exert neuroprotective effects. Specifically, it discusses how antioxidants, including polyphenols, vitamins, and flavonoids, contribute to the reduction in reactive oxygen species (ROS) production and neuroinflammation, thereby promoting neuronal survival and minimising oxidative damage in the brain. In addition, the article explores the role of antioxidants in modulating key molecular pathways involved in oxidative stress and neuroinflammation, such as the NF-κB, Nrf2, MAPK, and PI3K/AKT pathways, which regulate ROS generation, inflammatory cytokine expression, and antioxidant responses essential for maintaining cellular homeostasis in both the gut and the central nervous system. In addition, this review explores the complex relationship between gut-derived metabolites, oxidative stress, and neurodegenerative diseases, highlighting how dysbiosis-an imbalance in the gut microbiota-can exacerbate oxidative stress and contribute to neuroinflammation, thereby accelerating the progression of such diseases as AD and PD. The review also examines the role of short-chain fatty acids (SCFAs) produced by beneficial gut bacteria in modulating these pathways to attenuate neuroinflammation and oxidative damage. Furthermore, the article explores the therapeutic potential of microbiota-targeted interventions, including antioxidant delivery by probiotics and prebiotics, as innovative strategies to restore microbial homeostasis and support brain health. By synthesising current knowledge on the interplay between antioxidants, the gut-brain axis, and the molecular mechanisms underlying neurodegeneration, this review highlights the therapeutic promise of antioxidant-based interventions in mitigating oxidative stress and neurodegenerative disease progression. It also highlights the need for further research into antioxidant-rich dietary strategies and microbiota-focused therapies as promising avenues for the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Natalia Kurhaluk
- Institute of Biology, Pomeranian University in Słupsk, Arciszewski St. 22 B, 76-200 Słupsk, Poland;
| | - Piotr Kamiński
- Department of Medical Biology and Biochemistry, Division of Ecology and Environmental Protection, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, M. Skłodowska-Curie St. 9, 85-094 Bydgoszcz, Poland;
- Department of Biotechnology, Institute of Biological Sciences, Faculty of Biological Sciences, University of Zielona Góra, Prof. Z. Szafran St. 1, 65-516 Zielona Góra, Poland
| | - Rafał Bilski
- Department of Medical Biology and Biochemistry, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, M. Karłowicz St. 24, 85-092 Bydgoszcz, Poland; (R.B.); (R.K.); (A.W.)
| | - Renata Kołodziejska
- Department of Medical Biology and Biochemistry, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, M. Karłowicz St. 24, 85-092 Bydgoszcz, Poland; (R.B.); (R.K.); (A.W.)
| | - Alina Woźniak
- Department of Medical Biology and Biochemistry, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, M. Karłowicz St. 24, 85-092 Bydgoszcz, Poland; (R.B.); (R.K.); (A.W.)
| | - Halina Tkaczenko
- Institute of Biology, Pomeranian University in Słupsk, Arciszewski St. 22 B, 76-200 Słupsk, Poland;
| |
Collapse
|
21
|
Ibork H, Ait Lhaj Z, Boualam K, El Idrissi S, B Ortaakarsu A, Hajji L, Manalo Morgan A, Khallouki F, Taghzouti K, Abboussi O. Cannabidiol-Rich Cannabis sativa L. Extract Alleviates LPS-Induced Neuroinflammation Behavioral Alterations, and Astrocytic Bioenergetic Impairment in Male Mice. J Neurosci Res 2025; 103:e70035. [PMID: 40195769 DOI: 10.1002/jnr.70035] [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: 12/04/2024] [Revised: 02/25/2025] [Accepted: 03/25/2025] [Indexed: 04/09/2025]
Abstract
Neuroinflammation is a hallmark of various neurodegenerative disorders, yet effective treatments remain limited. This study investigates the neuroprotective potential of a cannabidiol (CBD)-Rich Cannabis sativa L. (CS) extract in a lipopolysaccharide (LPS)-induced neuroinflammation mouse model. The effects on anxiety-like behavior, cognitive function, and locomotor activity were assessed using behavioral tests (open field, elevated plus maze, novel object recognition, and Morris water maze). Antioxidant activity was measured by assaying glutathione (GSH) levels and lipid peroxidation by-products (TBARs). Anti-inflammatory properties were evaluated using quantitative reverse transcription polymerase chain reaction (QRt-PCR) for proinflammatory cytokines (IL-6 and TNF-α), glial fibrillary acidic protein (GFAP), and cannabinoid receptor 1 (CB1) mRNAs in the prefrontal cortex (PFC). Astrocytic bioenergetics were analyzed using extracellular flux assays. Additionally, computational inference with a deep learning approach was conducted to evaluate the synergistic interactions among CS phytocompounds on the CB1 receptors. Compared with synthetic CBD, the CS extract (20.0 mg/kg) demonstrated superior efficacy in mitigating LPS-induced anxiety-like behavior, cognitive deficits, and locomotor impairments. It also significantly mitigated oxidative stress (increased GSH, reduced TBARs) and suppressed proinflammatory cytokines and GFAP mRNAs, indicating potent anti-inflammatory properties. The extract modulated CB1 receptor expression and preserved metabolic homeostasis in cortical astrocytes, preventing their shift from glycolysis to oxidative phosphorylation under neuroinflammatory conditions. Computational modeling highlighted conformational changes in CB1 receptor residues induced by Delta-9-THC that enhanced CBD binding. These findings underscore the potential of CS extract as a therapeutic candidate for managing neuroinflammation and its associated neurodegenerative consequences, warranting further clinical exploration.
Collapse
Affiliation(s)
- Hind Ibork
- Physiology and Physiopathology Team, Faculty of Sciences, Genomic of Human Pathologies Research Centre, Mohammed V University in Rabat, Rabat, Morocco
| | - Zakaria Ait Lhaj
- Physiology and Physiopathology Team, Faculty of Sciences, Genomic of Human Pathologies Research Centre, Mohammed V University in Rabat, Rabat, Morocco
| | - Khadija Boualam
- Physiology and Physiopathology Team, Faculty of Sciences, Genomic of Human Pathologies Research Centre, Mohammed V University in Rabat, Rabat, Morocco
| | - Sara El Idrissi
- Physiology and Physiopathology Team, Faculty of Sciences, Genomic of Human Pathologies Research Centre, Mohammed V University in Rabat, Rabat, Morocco
| | - Ahmet B Ortaakarsu
- Department of Chemistry, Faculty of Science, Gazi University, Ankara, Turkey
| | - Lhoussain Hajji
- Bioactives, Health and Environmental Laboratory, Epigenetics Research Team, Moulay Ismail University, Meknes, Morocco
| | | | - Farid Khallouki
- Team of Ethnopharmacology and Pharmacognosy, Biology Department, Faculty of Sciences and Techniques Errachidia, Moulay Ismail University of Meknes, Errachidia, Morocco
| | - Khalid Taghzouti
- Physiology and Physiopathology Team, Faculty of Sciences, Genomic of Human Pathologies Research Centre, Mohammed V University in Rabat, Rabat, Morocco
| | - Oualid Abboussi
- Physiology and Physiopathology Team, Faculty of Sciences, Genomic of Human Pathologies Research Centre, Mohammed V University in Rabat, Rabat, Morocco
| |
Collapse
|
22
|
Chen X, Wu L, Lei Y, Tang H, Yan Z, Zhu S, Wen T, Zhu Z. A polysaccharide from Morchella esculenta mycelia: Structural characterization and protective effect on antioxidant stress on PC12 cells against H 2O 2-induced oxidative damage. Int J Biol Macromol 2025; 298:139886. [PMID: 39818380 DOI: 10.1016/j.ijbiomac.2025.139886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/24/2024] [Accepted: 01/13/2025] [Indexed: 01/18/2025]
Abstract
Morchella esculenta (L.) Pers. is considered a precious edible and medicinal fungus due to its strict growth environment requirements, difficult to cultivate, resulted in expensive in the market. A polysaccharide (MMP-L) was isolated from the mycelia of Morchella esculenta, with molecular weight of 3.02 × 103 kDa. MMP-L was composed of galactose, glucose, and mannose in a molar ratio of 1.00: 12.89: 0.29. Structural characterization showed that MMP-L had a backbone mainly consisting of →4)-α-D-Glcp-(1→, →6)-α-D-Glcp-(1→, →4,6)-α-D-Glcp-(1→, →2)-α-D-Galp-(1→, →3)-α-D-Manp-(1→, and →4,6)-α-D-Manp-(1→, and branched chains incorporating of α-D-Glcp(1→. The antioxidant activity test results indicated that MMP-L has effective scavenging ability against 1,1-diphenyl-2-picrylhydrazyl (DPPH), 2'2-Azinobis-(3-ethylbenzthiazoline-6-sulphonate) (ABTS), and hydroxyl radicals. Within the range of concentration (0.05-3.20 mg/mL), MMP-L increased the enzyme activity of superoxide dismutase (SOD) and glutathione (GSH) in H2O2-induced PC12 cells in a dose-dependent manner, while inhibiting the content of lactate dehydrogenase (LDH) and malondialdehyde (MDA). Furthermore, the decrease in reactive oxygen species (ROS) and apoptosis in PC12 cells may be attributed to the activation of the PI3K/Akt signaling pathway and downstream mitochondrial apoptosis-related protein Bcl-2/BAX/Caspase-3 signaling pathways. In summary, MMP-L might serve as a potential functional components for the future prevention and treatment of neurodegenerative disorders caused by oxidative damage.
Collapse
Affiliation(s)
- Xintao Chen
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin 300457, PR China; State Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology, Tianjin 300457, PR China; College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Lei Wu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin 300457, PR China; State Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology, Tianjin 300457, PR China; College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Yaru Lei
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin 300457, PR China; State Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology, Tianjin 300457, PR China; College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Hui Tang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin 300457, PR China; State Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology, Tianjin 300457, PR China; College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Zhejiang Yan
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin 300457, PR China; State Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology, Tianjin 300457, PR China; College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Sijie Zhu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin 300457, PR China; State Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology, Tianjin 300457, PR China; College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Tingchi Wen
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025,PR China; The Engineering Research Center of Southwest Bio-Pharmaceutical Resources, Ministry of Education, Guizhou University, Guiyang 550025, PR China
| | - Zhenyuan Zhu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin 300457, PR China; State Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology, Tianjin 300457, PR China; College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, PR China.
| |
Collapse
|
23
|
Raza A, Raina J, Sahu SK, Wadhwa P. Genetic mutations in kinases: a comprehensive review on marketed inhibitors and unexplored targets in Parkinson's disease. Neurol Sci 2025; 46:1509-1524. [PMID: 39760821 DOI: 10.1007/s10072-024-07970-2] [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: 10/15/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025]
Abstract
This comprehensive review navigates the landscape of genetic mutations in kinases, offering a thorough examination of both marketed inhibitors and unexplored targets in the context of Parkinson's Disease (PD). Although existing treatments for PD primarily center on symptom management, progress in comprehending the molecular foundations of the disease has opened avenues for targeted therapeutic approaches. This review encompasses an in-depth analysis of four key kinases-PINK1, LRRK2, GAK, and PRKRA-revealing that LRRK2 has garnered the most attention with a plethora of marketed inhibitors. However, the study underscores notable gaps in the exploration of inhibitors for PINK1, GAK, and a complete absence for PRKRA. The observed scarcity of inhibitors for these kinases emphasizes a significant area of untapped potential in PD therapeutics. By drawing attention to these unexplored targets, the review highlights the urgent need for focused research and drug development efforts to diversify the therapeutic landscape, potentially providing novel interventions for halting or slowing the progression of PD.
Collapse
Affiliation(s)
- Amir Raza
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar- Grand Trunk Rd, Phagwara, Punjab, India
| | - Jeevika Raina
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar- Grand Trunk Rd, Phagwara, Punjab, India
| | - Sanjeev Kumar Sahu
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar- Grand Trunk Rd, Phagwara, Punjab, India
| | - Pankaj Wadhwa
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar- Grand Trunk Rd, Phagwara, Punjab, India.
| |
Collapse
|
24
|
Nasb M, Li F, Dayoub L, Wu T, Wei M, Chen N. Bridging the gap: Integrating exercise mimicry into chronic disease management through suppressing chronic inflammation. J Adv Res 2025; 70:307-322. [PMID: 38704088 PMCID: PMC11976426 DOI: 10.1016/j.jare.2024.04.034] [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: 01/26/2024] [Revised: 03/25/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Chronic inflammation is a common hallmark of many chronic diseases. Although exercise holds paramount importance in preventing and managing chronic diseases, adherence to exercise programs can be challenging for some patients. Consequently, there is a pressing need to explore alternative strategies to emulate the anti-inflammatory effects of exercise for chronic diseases. AIM OF REVIEW This review explores the emerging role of green tea bioactive components as potential mitigators of chronic inflammation, offering insights into their capacity to mimic the beneficial effects of exercise. We propose that bioactive components in green tea are promising agents for suppressing chronic inflammation, suggesting their unique capability to replicate the health benefits of exercise. KEY SCIENTIFIC CONCEPTS OF REVIEW This review focuses on several key concepts, including chronic inflammation and its role in chronic diseases, the anti-inflammatory effects of regular exercise, and bioactive components in green tea responsible for its health benefits. It elaborates on scientific evidence supporting the anti-inflammatory properties of green tea bioactive components, such as epigallocatechin gallate (EGCG), and theorizes how these bioactive components might replicate the effects of exercise at a molecular level. Through a comprehensive analysis of current research, this review proposes a novel perspective on the application of green tea as a potential intervention strategy to suppress chronic inflammation, thereby extending the benefits akin to those achieved through exercise.
Collapse
Affiliation(s)
- Mohammad Nasb
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Fengxing Li
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Lamis Dayoub
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Tong Wu
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Minhui Wei
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Ning Chen
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China.
| |
Collapse
|
25
|
Adnan M, Siddiqui AJ, Bardakci F, Surti M, Badraoui R, Patel M. Mechanistic Insights into the Neuroprotective Potential of Aegle marmelos (L.) Correa Fruits against Aβ-Induced Cell Toxicity in Human Neuroblastoma SH-SY5Y Cells. Pharmaceuticals (Basel) 2025; 18:489. [PMID: 40283926 PMCID: PMC12030591 DOI: 10.3390/ph18040489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/20/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Amyloid-β (Aβ) plaque accumulation, oxidative stress, and cholinergic dysfunction are hallmarks of Alzheimer's disease (AD), a neurodegenerative disability that progresses over time, ultimately resulting in the loss of neurons. The side effects and limitations of current synthetic drugs have shifted attention toward natural alternatives. This study investigates the ethanolic extract of Aegle marmelos (L.) Corrêa fruits for their antioxidant, AChE-inhibitory, and anti-amyloidogenic properties, as well as their neuroprotective effects against amyloid beta-peptide (Aβ1-42). Methods: Phytochemical constituents were identified through HR-LCMS analysis and their antioxidant (DPPH, FRAP) and neuroprotective activities (AChE inhibition, ThT binding, MTT assay, ROS reduction, MMP restoration, and AD-related gene expression via qRT-PCR) were assessed using SHSY-5Y neuroblastoma cells. Results: The extract revealed the existence of flavonoids, phenols, and other bioactive substances. In vitro assays demonstrated strong antioxidant and AChE-inhibitory activities, while the ThT binding assay showed protection against amyloid-β aggregation. The extract exhibited no cytotoxicity in SHSY-5Y cells, even at a concentration of 500 μg/mL, whereas Aβ1-42 at 20 μM induced significant cytotoxicity. Co-treatment with Aβ1-42 (10 μM and 20 μM) and the extract improved cell viability (˃50%) and reduced ROS levels. Additionally, the extract restored mitochondrial membrane potential in Aβ1-42 treated cells, highlighting its role in preserving mitochondrial function. Conclusions: These findings suggest that A. marmelos fruits serve as a powerful source of natural antioxidants, AChE inhibitors, and anti-amyloidogenic agents, positioning them as a compelling option for AD treatment.
Collapse
Affiliation(s)
- Mohd Adnan
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (M.A.)
- King Salman Center for Disability Research, Riyadh 11614, Saudi Arabia
| | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (M.A.)
| | - Fevzi Bardakci
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (M.A.)
| | - Malvi Surti
- Research and Development Cell (RDC), Parul University, Waghodia, Vadodara 391760, Gujarat, India
- Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Waghodia, Vadodara 391760, Gujarat, India
| | - Riadh Badraoui
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (M.A.)
| | - Mitesh Patel
- Research and Development Cell (RDC), Parul University, Waghodia, Vadodara 391760, Gujarat, India
- Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Waghodia, Vadodara 391760, Gujarat, India
| |
Collapse
|
26
|
Wang H, Zhao YC, Xu L, Zhang TJ, Liu LH, Zhou MQ, Zhang H, Yang YN, Pan P, Jin L, Zhang ZW, Zhang XZ, Zhang LL. HIF-1α mediates mitochondrial damage by down-regulating ALKBH7 expression to promote the aberrant activation of FLS in rheumatoid arthritis. Acta Pharmacol Sin 2025:10.1038/s41401-025-01520-y. [PMID: 40140527 DOI: 10.1038/s41401-025-01520-y] [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: 06/22/2024] [Accepted: 02/23/2025] [Indexed: 03/28/2025]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovial inflammation and progressive joint destruction. Existing evidence indicates that hypoxia potentially contributes to the pathology of RA, though the specific mechanism remains unidentified. In this study, we explored the molecular mechanism through which the hypoxia-inducible factor (HIF-1α) contributed to the pathological process of RA. Our preliminary results suggested that hypoxia stimulates the activation of fibroblast-like synoviocytes (FLS) by inducing mitochondrial damage to activate cGAS-STING signaling, which can be effectively inhibited by silencing HIF-1α. In line with this, HIF-1α deficiency significantly alleviated the symptoms of collagen-induced arthritis (CIA) mice. RNA-Seq and CUT-Tag analysis revealed that HIF-1α down-regulated the expression of AlkB homologue 7 (ALKBH7) by acting on the ALKBH7 promoter site on chromosome 19 6372400-6372578. Using dual luciferase reporter analysis, we identified that ACCGTGGC as the motif to which HIF-1α bound directly. Subsequently, we demonstrated that knockdown of ALKBH7 induces mitochondrial damage and activates cGAS-STING signaling by downregulating the expression of UQCRC2. Conversely, overexpression of ALKBH7 could resist hypoxia-induced mitochondrial damage and FLS activation. In conclusion, HIF-1α triggers mitochondrial damage by downregulating the expression of ALKBH7 thereby promoting FLS activation, which may be the molecular mechanism by which hypoxia is involved in the pathological process of RA. Hypoxia promotes the activation of FLS through the induction of mitochondrial damage, which subsequently activates cGAS-STING signaling. Mechanistically, HIF-1α triggers mitochondrial damage by downregulating the expression of ALKBH7 in a target manner. Furthermore, the deletion of ALKBH7 leads to mitochondrial damage under hypoxic conditions, primarily through the downregulation of UQCRC2, as opposed to other complexes.
Collapse
Affiliation(s)
- Han Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China
| | - Yu-Chen Zhao
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China
| | - Li Xu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China
| | - Tian-Jing Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China
| | - Liang-Hu Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China
| | - Meng-Qi Zhou
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China
| | - Han Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China
| | - Yin-Ning Yang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China
| | - Pin Pan
- The Second People's Hospital of Hefei, Hefei, 230011, China
| | - Lin Jin
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China
| | - Zi-Wei Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China
| | - Xian-Zheng Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China.
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China.
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China.
- Department of Oncology, The First Affiliated Hospital, Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, China.
| | - Ling-Ling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China.
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China.
- Anti-inflammatory Immune Drugs Collaborative Innovation Center, Anhui Province, Hefei, 230032, China.
| |
Collapse
|
27
|
Dehghani S, Ocakcı O, Hatipoglu PT, Özalp VC, Tevlek A. Exosomes as Biomarkers and Therapeutic Agents in Neurodegenerative Diseases: Current Insights and Future Directions. Mol Neurobiol 2025:10.1007/s12035-025-04825-5. [PMID: 40095345 DOI: 10.1007/s12035-025-04825-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/07/2025] [Indexed: 03/19/2025]
Abstract
Neurodegenerative diseases (NDs) like Alzheimer's, Parkinson's, and ALS rank among the most challenging global health issues, marked by substantial obstacles in early diagnosis and effective treatment. Current diagnostic techniques frequently demonstrate inadequate sensitivity and specificity, whilst conventional treatment strategies encounter challenges related to restricted bioavailability and insufficient blood-brain barrier (BBB) permeability. Recently, exosomes-nanoscale vesicles packed with proteins, RNAs, and lipids-have emerged as promising agents with the potential to reshape diagnostic and therapeutic approaches to these diseases. Unlike conventional drug carriers, they naturally traverse the BBB and can deliver bioactive molecules to affected neural cells. Their molecular cargo can influence cell signaling, reduce neuroinflammation, and potentially slow neurodegenerative progression. Moreover, exosomes serve as non-invasive biomarkers, enabling early and precise diagnosis while allowing real-time disease monitoring. Additionally, engineered exosomes, loaded with therapeutic molecules, enhance this capability by targeting diseased neurons and overcoming conventional treatment barriers. By offering enhanced specificity, reduced immunogenicity, and an ability to bypass physiological limitations, exosome-based strategies present a transformative advantage over existing diagnostic and therapeutic approaches. This review examines the multifaceted role of exosomes in NDDs, emphasizing their diagnostic capabilities, intrinsic therapeutic functions, and transformative potential as advanced treatment vehicles.
Collapse
Affiliation(s)
- Sam Dehghani
- Faculty of Medicine, Undergraduate Program, Atılım University, 06830, Ankara, Turkey
| | - Ozgecan Ocakcı
- Department of Medical Biology, Faculty of Medicine, AtıLıM University, 06830, Ankara, Turkey
| | - Pars Tan Hatipoglu
- Faculty of Medicine, Undergraduate Program, Atılım University, 06830, Ankara, Turkey
| | - Veli Cengiz Özalp
- Department of Medical Biology, Faculty of Medicine, AtıLıM University, 06830, Ankara, Turkey
| | - Atakan Tevlek
- Department of Medical Biology, Faculty of Medicine, AtıLıM University, 06830, Ankara, Turkey.
| |
Collapse
|
28
|
Caturano A, Erul E, Nilo R, Nilo D, Russo V, Rinaldi L, Acierno C, Gemelli M, Ricotta R, Sasso FC, Giordano A, Conte C, Ürün Y. Insulin resistance and cancer: molecular links and clinical perspectives. Mol Cell Biochem 2025:10.1007/s11010-025-05245-8. [PMID: 40089612 DOI: 10.1007/s11010-025-05245-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/23/2025] [Indexed: 03/17/2025]
Abstract
The association between insulin resistance (IR), type 2 diabetes mellitus (T2DM), and cancer is increasingly recognized and poses an escalating global health challenge, as the incidence of these conditions continues to rise. Studies indicate that individuals with T2DM have a 10-20% increased risk of developing various solid tumors, including colorectal, breast, pancreatic, and liver cancers. The relative risk (RR) varies depending on cancer type, with pancreatic and liver cancers showing a particularly strong association (RR 2.0-2.5), while colorectal and breast cancers demonstrate a moderate increase (RR 1.2-1.5). Understanding these epidemiological trends is crucial for developing integrated management strategies. Given the global rise in T2DM and cancer cases, exploring the complex relationship between these conditions is critical. IR contributes to hyperglycemia, chronic inflammation, and altered lipid metabolism. Together, these factors create a pro-tumorigenic environment conducive to cancer development and progression. In individuals with IR, hyperinsulinemia triggers the insulin-insulin-like growth factor (IGF1R) signaling pathway, activating cancer-associated pathways such as mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PIK3CA), which promote cell proliferation and survival, thereby supporting tumor growth. Both IR and T2DM are linked to increased morbidity and mortality in patients with cancer. By providing an in-depth analysis of the molecular links between insulin resistance and cancer, this review offers valuable insights into the role of metabolic dysfunction in tumor progression. Addressing insulin resistance as a co-morbidity may open new avenues for risk assessment, early intervention, and the development of integrated treatment strategies to improve patient outcomes.
Collapse
Affiliation(s)
- Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138, Naples, Italy
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166, Rome, Italy
| | - Enes Erul
- Department of Medical Oncology, Faculty of Medicine, Ankara University, Ankara, 06620, Turkey
| | - Roberto Nilo
- Data Collection G-STeP Research Core Facility, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - Davide Nilo
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138, Naples, Italy
| | - Vincenzo Russo
- Department of Biology, College of Science and Technology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, 19122, USA
- Division of Cardiology, Department of Medical Translational Sciences, University of Campania Luigi Vanvitelli, 80138, Naples, Italy
| | - Luca Rinaldi
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, 86100, Campobasso, Italy
| | - Carlo Acierno
- Azienda Ospedaliera Regionale San Carlo, 85100, Potenza, Italy
| | - Maria Gemelli
- Medical Oncology Unit, IRCCS MultiMedica, Milan, Italy
| | | | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138, Naples, Italy
| | - Antonio Giordano
- Department of Biology, College of Science and Technology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, 19122, USA
| | - Caterina Conte
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166, Rome, Italy
- Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, 20099, Milan, Italy
| | - Yüksel Ürün
- Department of Medical Oncology, Faculty of Medicine, Ankara University, Ankara, 06620, Turkey.
| |
Collapse
|
29
|
Daubermann C, Herhaus B, Neuberger EWI, Simon P, Petrowski K. Methodological influences on circulating cell-free-mitochondrial and nuclear DNA concentrations in response to chronic stress. Mol Biol Rep 2025; 52:303. [PMID: 40080226 PMCID: PMC11906544 DOI: 10.1007/s11033-025-10369-7] [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: 12/18/2024] [Accepted: 02/14/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Mitochondria are versatile eukaryotic organelles that play a crucial role in the body's stress response. Prolonged stress exposure can cause structural and functional alterations, leading to mitochondrial DNA (mtDNA) damage and subsequent release of mtDNA into the circulation. Cell-free circulating mtDNA (ccf-mtDNA) is a potential biomarker indicating cellular damage and stress. In this study we investigated the applicability of ccf-mtDNA and cf-nDNA as biomarkers of chronic stress in healthy subjects. METHODS AND RESULTS We developed a quantitative polymerase chain reaction (qPCR) assay to directly measure ccf-mtDNA in human blood plasma samples, addressing numerous challenges specifically related to ccf-mtDNA quantification. We validated our 68 bp target assay based on the FDA, International Organization for Standardization (ISO) and Clinical & Laboratory Standards Institute (CLSI) guidelines for assay development, including parameters such as limit of blank (LOB), limit of detection (LOD) and limit of quantification (LOQ). Furthermore, we implemented incurred samples analysis and inter-plate samples to ensure reliability and reproducibility of the assay. In addition, we evaluated the effects of centrifugation forces on ccf-mtDNA and cf-nDNA concentrations in native plasma samples and showed that mainly ccf-mtDNA is strongly affected by centrifugation forces. We found a significant negative correlation between ccf-mtDNA levels and chronic stress. In contrast, cf-nDNA levels were not affected in response to chronic stress. CONCLUSION ccf-mtDNA can directly and reliably quantified in unpurified plasma samples. However, the ccf-mtDNA levels in plasma samples of healthy subjects are close the LOQ, showing that the assay is not yet suitable for all conditions.
Collapse
Affiliation(s)
- Carina Daubermann
- Department of Sports Medicine, Rehabilitation and Disease Prevention, Johannes Gutenberg University Mainz, Albert-Schweitzer Strasse 22, 55128, Mainz, Germany
| | - Benedict Herhaus
- Department of Medical Psychology and Medical Sociology, University Medical Centre of the Johannes Gutenberg-University, Duesbergweg 6, 55128, Mainz, Germany
| | - Elmo W I Neuberger
- Department of Sports Medicine, Rehabilitation and Disease Prevention, Johannes Gutenberg University Mainz, Albert-Schweitzer Strasse 22, 55128, Mainz, Germany
| | - Perikles Simon
- Department of Sports Medicine, Rehabilitation and Disease Prevention, Johannes Gutenberg University Mainz, Albert-Schweitzer Strasse 22, 55128, Mainz, Germany.
| | - Katja Petrowski
- Department of Medical Psychology and Medical Sociology, University Medical Centre of the Johannes Gutenberg-University, Duesbergweg 6, 55128, Mainz, Germany.
| |
Collapse
|
30
|
Hermann DM, Wang C, Mohamud Yusuf A, Herz J, Doeppner TR, Giebel B. Extracellular vesicles lay the ground for neuronal plasticity by restoring mitochondrial function, cell metabolism and immune balance. J Cereb Blood Flow Metab 2025:271678X251325039. [PMID: 40072028 PMCID: PMC11904928 DOI: 10.1177/0271678x251325039] [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: 09/14/2024] [Revised: 01/15/2025] [Accepted: 02/14/2025] [Indexed: 03/15/2025]
Abstract
Extracellular vesicles (EVs) convey complex signals between cells that can be used to promote neuronal plasticity and neurological recovery in brain disease models. These EV signals are multimodal and context-dependent, making them unique therapeutic principles. This review analyzes how EVs released from various cell sources control neuronal metabolic function, neuronal survival and plasticity. Preferential sites of EV communication in the brain are interfaces between pre- and postsynaptic neurons at synapses, between astrocytes and neurons at plasma membranes or tripartite synapses, between oligodendrocytes and neurons at axons, between microglial cells/macrophages and neurons, and between cerebral microvascular cells and neurons. At each of these interfaces, EVs support mitochondrial function and cell metabolism under physiological conditions and orchestrate neuronal survival and plasticity in response to brain injury. In the injured brain, the promotion of neuronal survival and plasticity by EVs is tightly linked with EV actions on mitochondrial function, cell metabolism, oxidative stress and immune responses. Via the stabilization of cell metabolism and immune balance, neuronal plasticity responses are activated and functional neurological recovery is induced. As such, EV lay the ground for neuronal plasticity.
Collapse
Affiliation(s)
- Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Chen Wang
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ayan Mohamud Yusuf
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Josephine Herz
- Department of Pediatrics I, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Thorsten R Doeppner
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bernd Giebel
- Department of Neurology, University Hospital Gießen and Marburg, Justus-Liebig-University Gießen, Gießen, Germany
| |
Collapse
|
31
|
Wang J, Gu D, Jin K, Shen H, Qian Y. The Role of G-Protein-Coupled Receptor Kinase 4 in Modulating Mitophagy and Oxidative Stress in Cerebral Ischemia-Reperfusion Injury. Neuromolecular Med 2025; 27:21. [PMID: 40055267 DOI: 10.1007/s12017-025-08843-3] [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: 01/28/2025] [Accepted: 02/22/2025] [Indexed: 05/13/2025]
Abstract
Cerebral ischemia-reperfusion injury (CIRI) causes significant neuronal damage through oxidative stress, inflammation, and mitochondrial dysfunction. The G-protein-coupled receptor kinase 4 (GRK4) has been implicated in regulating stress responses in various tissues, but its role in ischemic brain injury remains unclear. In this study, we investigated the role of GRK4 in oxidative stress, inflammation, and mitophagy during CIRI using both in vivo and in vitro models. For the in vivo experiments, we employed the bilateral common carotid artery occlusion (BCCAO) model to induce ischemia-reperfusion injury. Our finding demonstrated that ischemic reperfusion significantly upregulated GRK4 expression in the brain, correlating with elevated levels of inflammatory cytokines and oxidative stress markers. In cultured cerebellar neurons subjected to oxygen-glucose deprivation (OGD), over-expression of GRK4 decreased cell viability, while GRK4 inhibition enhanced neuronal survival, suggesting that GRK4 exacerbates neuronal damage in ischemic conditions. Furthermore, GRK4 overexpression impaired mitophagy, as indicated by altered expression of key mitophagy-related proteins (Beclin-1, PINK1, and p62), which led to mitochondrial dysfunction and increased oxidative stress. In contrast, GRK4 inhibition promoted more efficient mitophagy and improved mitochondrial quality control. These results highlight the detrimental role of GRK4 in ischemic brain injury and suggest that targeting GRK4 could offer a novel therapeutic strategy to mitigate neuronal damage by balancing oxidative stress, inflammation, and mitochondrial dynamics. Further studies are needed to elucidate the precise molecular mechanisms underlying GRK4-mediated neuroinflammation and mitochondrial dysfunction in ischemic stroke.
Collapse
Affiliation(s)
- Jian Wang
- Department of Neurosurgery, Taicang Hospital of Traditional Chinese Medicine, Affiliated to Nanjing University of Traditional Chinese Medicine, Suzhou, 215400, Jiangsu, China.
| | - Diheng Gu
- Department of Neurosurgery, Taicang Hospital of Traditional Chinese Medicine, Affiliated to Nanjing University of Traditional Chinese Medicine, Suzhou, 215400, Jiangsu, China
| | - Ke Jin
- Department of Neurosurgery, Taicang Hospital of Traditional Chinese Medicine, Affiliated to Nanjing University of Traditional Chinese Medicine, Suzhou, 215400, Jiangsu, China
| | - Hualong Shen
- Department of Neurosurgery, Taicang Hospital of Traditional Chinese Medicine, Affiliated to Nanjing University of Traditional Chinese Medicine, Suzhou, 215400, Jiangsu, China
| | - Yaohua Qian
- Department of Neurosurgery, Taicang Hospital of Traditional Chinese Medicine, Affiliated to Nanjing University of Traditional Chinese Medicine, Suzhou, 215400, Jiangsu, China
| |
Collapse
|
32
|
Nakamya MF, Hu K, Jiang C, Chong Z, Liu RM. Age- and ApoE Genotype-Dependent Transcriptomic Responses to O 3 in the Hippocampus of Mice. Int J Mol Sci 2025; 26:2407. [PMID: 40141051 PMCID: PMC11942628 DOI: 10.3390/ijms26062407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/25/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
Alzheimer's disease (AD) is a leading cause of dementia in the elderly, with late-onset AD (LOAD) accounting for 95% of the cases. The etiology underlying LOAD, however, remains unclear. Using a humanized mouse model, we showed previously that exposure to ozone (O3), a potential environment risk factor, in a cyclic exposure protocol that mimics a human exposure scenario, accelerated AD-like neuropathophysiology in old humanized male ApoE3 (E3) but not ApoE4 (E4) mice. Using RNA sequencing (RNA-seq) techniques, we further demonstrate here that the ApoE genotype has the greatest influence on transcriptional changes, followed by age and O3 exposure. Notably, AD-related genes were expressed even at baseline and in young mice, but the differences in the expression levels are obvious in old age. Importantly, although both E3 and E4 mice exhibited some AD-related transcriptomic alterations, old E3 mice exposed to O3, which showed memory impairment, experienced more pronounced disruptions in the expression of genes related to redox balance, neurogenesis, neuroinflammation, and cellular senescence in the hippocampus, compared with O3-exposed old E4 mice. These results provide new insights into the molecular mechanisms underlying memory loss in O3-exposed old E3 male mice and emphasize the complexity of interactions between gene, environment, and aging in AD pathophysiology.
Collapse
Affiliation(s)
- Mary F. Nakamya
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (M.F.N.); (C.J.)
| | - Kaili Hu
- Department of Biomedical Informatics and Data Science, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Chunsun Jiang
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (M.F.N.); (C.J.)
| | - Zechen Chong
- Department of Biomedical Informatics and Data Science, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Rui-Ming Liu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (M.F.N.); (C.J.)
| |
Collapse
|
33
|
Fu C, Weng S, Liu D, Guo R, Chen M, Shi B, Weng J. Review on the Role of Mitochondrial Dysfunction in Septic Encephalopathy. Cell Biochem Biophys 2025; 83:135-145. [PMID: 39212823 DOI: 10.1007/s12013-024-01493-5] [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] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Septic Encephalopathy (SE) is a frequent and severe complication of sepsis, characterized by a range of neurocognitive impairments from mild confusion to deep coma. The underlying pathophysiology of SE involves systemic inflammation, neuroinflammation, blood-brain barrier (BBB) disruption, and mitochondrial dysfunction. Among these factors, mitochondrial dysfunction plays a pivotal role, contributing to impaired ATP production, increased reactive oxygen species (ROS) generation, and activation of apoptotic pathways, all of which exacerbate neuronal damage and cognitive deficits. Diagnosis of SE relies on clinical evaluation, neuroimaging, electroencephalography (EEG), and laboratory tests, though specific diagnostic markers are still lacking. Epidemiological data show SE is prevalent in intensive care unit (ICU) patients, especially those with severe sepsis or septic shock, with incidence rates varying widely depending on the population and diagnostic criteria used. Recent research highlights the importance of mitochondrial dynamics, including biogenesis, fission, and fusion, in the development of SE. Mitophagy, a selective form of autophagy that degrades damaged mitochondria, plays a critical role in maintaining mitochondrial health and protecting against dysfunction. Targeting mitochondrial pathways and enhancing mitophagy offers a promising therapeutic strategy to mitigate the effects of SE, reduce oxidative stress, prevent apoptosis, and support the resolution of neuroinflammation. Further research is essential to elucidate the mechanisms of mitochondrial dysfunction and mitophagy in SE and develop effective interventions to improve patient outcomes.
Collapse
Affiliation(s)
- Chunjin Fu
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China
| | - Shuoyun Weng
- School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, 325035, China
| | - Danjuan Liu
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China
| | - Rongjie Guo
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China
| | - Min Chen
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China
| | - Bingbing Shi
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China
| | - Junting Weng
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China.
| |
Collapse
|
34
|
Munteanu C, Galaction AI, Onose G, Turnea M, Rotariu M. Harnessing Gasotransmitters to Combat Age-Related Oxidative Stress in Smooth Muscle and Endothelial Cells. Pharmaceuticals (Basel) 2025; 18:344. [PMID: 40143122 PMCID: PMC11946800 DOI: 10.3390/ph18030344] [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: 01/25/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Age-related oxidative stress is a critical factor in vascular dysfunction, contributing to hypertension and atherosclerosis. Smooth muscle cells and endothelial cells are particularly susceptible to oxidative damage, which exacerbates vascular aging through cellular senescence, chronic inflammation, and arterial stiffness. Gasotransmitters-hydrogen sulfide (H2S), nitric oxide (NO), and carbon monoxide (CO)-are emerging as promising therapeutic agents for counteracting these processes. This review synthesizes findings from recent studies focusing on the mechanisms by which H2S, NO, and CO influence vascular smooth muscle and endothelial cell function. Therapeutic strategies involving exogenous gasotransmitter delivery systems and combination therapies were analyzed. H2S enhances mitochondrial bioenergetics, scavenges ROS, and activates antioxidant pathways. NO improves endothelial function, promotes vasodilation, and inhibits platelet aggregation. CO exhibits cytoprotective and anti-inflammatory effects by modulating heme oxygenase activity and ROS production. In preclinical studies, gasotransmitter-releasing molecules (e.g., NaHS, SNAP, CORMs) and targeted delivery systems show significant promise. Synergistic effects with lifestyle modifications and antioxidant therapies further enhance their therapeutic potential. In conclusion, gasotransmitters hold significant promise as therapeutic agents to combat age-related oxidative stress in vascular cells. Their multifaceted mechanisms and innovative delivery approaches make them potential candidates for treating vascular dysfunction and promoting healthy vascular aging. Further research is needed to translate these findings into clinical applications.
Collapse
Affiliation(s)
- Constantin Munteanu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
| | - Anca Irina Galaction
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| | - Gelu Onose
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Marius Turnea
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| | - Mariana Rotariu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| |
Collapse
|
35
|
Jia H, Song Y, Hua Y, Li K, Li S, Wang Y. Molecular Mechanism of Aerobic Exercise Ameliorating Myocardial Mitochondrial Injury in Mice with Heart Failure. Int J Mol Sci 2025; 26:2136. [PMID: 40076760 PMCID: PMC11901053 DOI: 10.3390/ijms26052136] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/02/2024] [Accepted: 12/11/2024] [Indexed: 03/14/2025] Open
Abstract
To explore the molecular mechanism of aerobic exercise to improve heart failure and to provide a theoretical basis and experimental reference for the treatment of heart failure. Nine-week-old male mice were used to establish a left ventricular pressure overload-induced heart failure model by transverse aortic constriction (TAC). The mice were randomly divided into four groups: a sham group (SHAM), heart failure group (HF), heart failure + SKQ1 group (HS) and heart failure + aerobic exercise group (HE). The mice in the HE group were subjected to moderate-intensity aerobic exercise interventions. The mitochondrion-targeting antioxidant (SKQ1) contains the lipophilic cation TPP, which targets scavenging mitochondrial ROS. The HS group was subjected to SKQ1 (100 nmol/kg/d) interventions, which were initiated 1 week after the surgery, and the interventions lasted 8 weeks. Cardiac function was assessed by ultrasound, cardiomyocyte size by H&E and WGA staining, myocardial fibrosis by Masson's staining, and myocardial tissue oxidative stress and apoptosis by DHE and TUNEL fluorescence staining, respectively. Western blotting was used to detect the expression of mitochondrial quality control, inflammation, and apoptosis-related proteins. In the cellular level, an in vitro cellular model was established by isolating primary cardiomyocytes from neonatal mice (2-3 days) and intervening with Ang II (1 μM) to mimic heart failure. Oxidative stress and mitochondrial membrane potential were determined in the cardiomyocytes of each group by DHE and JC-1 staining, respectively. Myocardial fibrosis was increased significantly and cardiac function was reduced significantly in the heart failure mice. Aerobic exercise and SKQ1 intervention improved cardiac function and reduced myocardial hypertrophy and myocardial fibrosis in the heart failure mice significantly. Meanwhile, aerobic exercise and SKQ1 intervention reduced the number of DHE-positive particles (p < 0.01) and inhibited myocardial oxidative stress in the heart failure mice significantly. Aerobic exercise also reduced DRP1, Parkin, and BNIP3 protein expression (p < 0.05, p < 0.01), and increased OPA1 and PINK1 protein expression (p < 0.05, p < 0.01) significantly. Moreover, aerobic exercise and SKQ1 intervention decreased the number of TUNEL-positive particles and the expression of inflammation- and apoptosis-related proteins NLRP3, TXNIP, Caspase-1, IL-1β, BAX, BAK, and p53 significantly (p < 0.05, p < 0.01). In addition, the AMPK agonist AICAR and the mitochondria-targeted ROS scavenger (SKQ1) ameliorated AngII-induced mitochondrial fragmentation and decreased mitochondrial membrane potential in cardiomyocytes significantly. It was shown that inhibition of mitochondrial ROS by aerobic exercise, which in turn inhibits mitochondrial damage, improves mitochondrial quality control, and reduces myocardial inflammatory and apoptosis, may be an important molecular mechanism by which aerobic exercise exerts endogenous antioxidant protective effects to improve cardiac function.
Collapse
Affiliation(s)
| | | | | | | | | | - Youhua Wang
- Institute of Sports and Exercise Biology, School of Physical Education, Shanxi Normal University, Xi’an 710119, China; (H.J.); (Y.S.); (Y.H.); (K.L.); (S.L.)
| |
Collapse
|
36
|
Shirvani P, Shirvani A, Holick MF. Mitochondrial Dysfunction and Its Potential Molecular Interplay in Hypermobile Ehlers-Danlos Syndrome: A Scoping Review Bridging Cellular Energetics and Genetic Pathways. Curr Issues Mol Biol 2025; 47:134. [PMID: 39996855 PMCID: PMC11854588 DOI: 10.3390/cimb47020134] [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: 01/08/2025] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
Hypermobile Ehlers-Danlos Syndrome (hEDS) is a hereditary connective tissue disorder characterized by joint hypermobility, skin hyperextensibility, and systemic manifestations such as chronic fatigue, gastrointestinal dysfunction, and neurological symptoms. Unlike other EDS subtypes with known genetic mutations, hEDS lacks definitive markers, suggesting a multifactorial etiology involving both mitochondrial dysfunction and non-mitochondrial pathways. This scoping review, conducted in accordance with the PRISMA-ScR guidelines, highlights mitochondrial dysfunction as a potential unifying mechanism in hEDS pathophysiology. Impaired oxidative phosphorylation (OXPHOS), elevated reactive oxygen species (ROS) levels, and calcium dysregulation disrupt cellular energetics and extracellular matrix (ECM) homeostasis, contributing to the hallmark features of hEDS. We reviewed candidate genes associated with ECM remodeling, signaling pathways, and immune regulation. Protein-protein interaction (PPI) network analyses revealed interconnected pathways linking mitochondrial dysfunction with these candidate genes. Comparative insights from Fabry disease and fragile X premutation carriers underscore shared mechanisms such as RNA toxicity, matrix metalloproteinases (MMP) activation, and ECM degradation. These findings may suggest that mitochondrial dysfunction amplifies systemic manifestations through its interplay with non-mitochondrial molecular pathways. By integrating these perspectives, this review provides a potential framework for understanding hEDS pathogenesis while highlighting latent avenues for future research into its molecular basis. Understanding the potential role of mitochondrial dysfunction in hEDS not only sheds light on its complex molecular etiology but also opens new paths for targeted interventions.
Collapse
Affiliation(s)
| | - Arash Shirvani
- Ehlers-Danlos Syndrome Clinical Research Program, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Michael F. Holick
- Ehlers-Danlos Syndrome Clinical Research Program, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
| |
Collapse
|
37
|
Thawabteh AM, Ghanem AW, AbuMadi S, Thaher D, Jaghama W, Karaman D, Karaman R. Promising Natural Remedies for Alzheimer's Disease Therapy. Molecules 2025; 30:922. [PMID: 40005231 PMCID: PMC11858286 DOI: 10.3390/molecules30040922] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/04/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
This study examines the intricacies of Alzheimer's disease (AD), its origins, and the potential advantages of various herbal extracts and natural compounds for enhancing memory and cognitive performance. Future studies into AD treatments are encouraged by the review's demonstration of the effectiveness of phytoconstituents that were extracted from a number of plants. In addition to having many beneficial effects, such as improved cholinergic and cognitive function, herbal medicines are also much less harmful, more readily available, and easier to use than other treatments. They also pass without difficulty through the blood-brain barrier (BBB). This study focused on natural substances and their effects on AD by using academic databases to identify peer-reviewed studies published between 2015 and 2024. According to the literature review, 66 phytoconstituents that were isolated from 21 distinct plants have shown efficacy, which could be encouraging for future research on AD therapies. Since most clinical trials produce contradictory results, the study suggests that larger-scale studies with longer treatment durations are necessary to validate or refute the therapeutic efficacy of herbal AD treatments.
Collapse
Affiliation(s)
- Amin Mahmood Thawabteh
- Department of Chemistry, Birzeit University, West Bank, Ramallah 00972, Palestine;
- Faculty of Pharmacy, Nursing and Health Professions, Birzeit University, West Bank, Ramallah 00972, Palestine; (A.W.G.); (S.A.); (D.T.); (W.J.)
| | - Aseel Wasel Ghanem
- Faculty of Pharmacy, Nursing and Health Professions, Birzeit University, West Bank, Ramallah 00972, Palestine; (A.W.G.); (S.A.); (D.T.); (W.J.)
| | - Sara AbuMadi
- Faculty of Pharmacy, Nursing and Health Professions, Birzeit University, West Bank, Ramallah 00972, Palestine; (A.W.G.); (S.A.); (D.T.); (W.J.)
| | - Dania Thaher
- Faculty of Pharmacy, Nursing and Health Professions, Birzeit University, West Bank, Ramallah 00972, Palestine; (A.W.G.); (S.A.); (D.T.); (W.J.)
| | - Weam Jaghama
- Faculty of Pharmacy, Nursing and Health Professions, Birzeit University, West Bank, Ramallah 00972, Palestine; (A.W.G.); (S.A.); (D.T.); (W.J.)
| | - Donia Karaman
- Pharmaceutical Sciences Department, Faculty of Pharmacy, Al-Quds University, Jerusalem 20002, Palestine;
| | - Rafik Karaman
- Pharmaceutical Sciences Department, Faculty of Pharmacy, Al-Quds University, Jerusalem 20002, Palestine;
- Department of Sciences, University of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy
| |
Collapse
|
38
|
Yeganeh Markid T, Pourahmadiyan A, Hamzeh S, Sharifi-Bonab M, Asadi MR, Jalaiei A, Rezazadeh M, Ghafouri-Fard S. A special focus on polyadenylation and alternative polyadenylation in neurodegenerative diseases: A systematic review. J Neurochem 2025; 169:e16255. [PMID: 39556113 DOI: 10.1111/jnc.16255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 11/19/2024]
Abstract
Neurodegenerative diseases (NDDs) are one of the prevailing conditions characterized by progressive neuronal loss. Polyadenylation (PA) and alternative polyadenylation (APA) are the two main post-transcriptional events that regulate neuronal gene expression and protein production. This systematic review analyzed the available literature on the role of PA and APA in NDDs, with an emphasis on their contributions to disease development. A comprehensive literature search was performed using the PubMed, Scopus, Cochrane, Google Scholar, Embase, Web of Science, and ProQuest databases. The search strategy was developed based on the framework introduced by Arksey and O'Malley and supplemented by the inclusion and exclusion criteria. The study selection was performed by two independent reviewers. Extraction and data organization were performed in accordance with the predefined variables. Subsequently, quantitative and qualitative analyses were performed. Forty-seven studies were included, related to a variety of NDDs, namely Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. Disease induction was performed using different models, including human tissues, animal models, and cultured cells. Most investigations were related to PA, although some were related to APA or both. Amyloid precursor protein (APP), Tau, SNCA, and STMN2 were the major genes identified; most of the altered PA patterns were related to mRNA stability and translation efficiency. This review particularly underscores the key roles of PA and APA in the pathogenesis of NDDs through their mechanisms that contribute to gene expression dysregulation, protein aggregation, and neuronal dysfunction. Insights into these mechanisms may lead to new therapeutic strategies focused on the modulation of PA and APA activities. Further research is required to investigate the translational potential of targeting these pathways for NDD treatment.
Collapse
Affiliation(s)
- Tarlan Yeganeh Markid
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Azam Pourahmadiyan
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Soroosh Hamzeh
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mirmohsen Sharifi-Bonab
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohamad Reza Asadi
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Jalaiei
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Rezazadeh
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
39
|
Wang T, Huang X, Sun S, Wang Y, Han L, Zhang T, Zhang T, Chen X. Recent Advances in the Mechanisms of Postoperative Neurocognitive Dysfunction: A Narrative Review. Biomedicines 2025; 13:115. [PMID: 39857699 PMCID: PMC11762480 DOI: 10.3390/biomedicines13010115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/28/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Postoperative neurocognitive dysfunction (PND) is a prevalent and debilitating complication in elderly surgical patients, characterized by persistent cognitive decline that negatively affects recovery and quality of life. As the aging population grows, the rising number of elderly surgical patients has made PND an urgent clinical challenge. Despite increasing research efforts, the pathophysiological mechanisms underlying PND remain inadequately characterized, underscoring the need for a more integrated framework to guide targeted interventions. To better understand the molecular mechanisms and therapeutic targets of PND, this narrative review synthesized evidence from peer-reviewed studies, identified through comprehensive searches of PubMed, Embase, Cochrane Library, and Web of Science. Key findings highlight neuroinflammation, oxidative stress, mitochondrial dysfunction, neurotransmitter imbalances, microvascular changes, and white matter lesions as central to PND pathophysiology, with particular parallels to encephalocele- and sepsis-associated cognitive impairments. Among these, neuroinflammation, mediated by pathways such as the NLRP3 inflammasome and blood-brain barrier disruption, emerges as a pivotal driver, triggering cascades that exacerbate neuronal injury. Oxidative stress and mitochondrial dysfunction synergistically amplify these effects, while neurotransmitter imbalances and microvascular alterations, including white matter lesions, contribute to synaptic dysfunction and cognitive decline. Anesthetic agents modulate these interconnected pathways, exhibiting both protective and detrimental effects. Propofol and dexmedetomidine demonstrate neuroprotective properties by suppressing neuroinflammation and microglial activation, whereas inhalational anesthetics like sevoflurane intensify oxidative stress and inflammatory responses. Ketamine, with its anti-inflammatory potential, offers promise but requires further evaluation to determine its long-term safety and efficacy. By bridging molecular insights with clinical practice, this review highlights the critical role of personalized anesthetic strategies in mitigating PND and improving cognitive recovery in elderly surgical patients. It aims to inform future research and clinical decision-making to address this multifaceted challenge.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Xin Huang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Shujun Sun
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Yafeng Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Linlin Han
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Tao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Tianhao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| |
Collapse
|
40
|
Tang L, Li M, Piao S, Du L, Qiu S, Jiang X, Luo M, Wang Y, Pan Z. Activation of the Keap1/Nrf2/HO-1 Pathway by "Tianyu" Pairing: Implications for Inflammation and Oxidative Stress in Rheumatoid Arthritis. Endocr Metab Immune Disord Drug Targets 2025; 25:479-491. [PMID: 39192656 DOI: 10.2174/0118715303307608240812114651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/17/2024] [Accepted: 06/04/2024] [Indexed: 08/29/2024]
Abstract
OBJECTIVE The objective of this study was to examine the impact of "Tianyu" Pairing on oxidative stress in the development of Rheumatoid arthritis (RA) and approach its potential mechanism using cell experiments. METHODS A cell model of RA was developed by stimulating rheumatoid arthritis fibroblast-like synoviocytes (RA-FLS) with tumor necrosis factor-α (TNF-α). This model aimed to assess the impact of serum containing Rhodiola rosea-Euonymus alatus drug pair (TYP) on inflammation and oxidative stress in the development of RA, specifically through the Keap1/Nrf2/HO-1 pathway. RESULTS The findings from the in vitro experiment demonstrated that the presence of TYP in the serum effectively suppressed the proliferation of RA-FLS induced by TNF-α. Additionally, TYP facilitated the apoptosis of afflicted cells, attenuated the migratory and invasive capabilities of diseased cells, and decreased the levels of Kelch ECH associating protein 1 (Keap1), reactive oxygen species (ROS), glutathione peroxidase (GSH-Px), catalase (CAT), and malondialdehyde (MDA) (p < 0.01). The influence of inflammation and oxidative stress in RA-FLS cells was reduced by increasing the nuclear-cytoplasmic ratio of Nuclear Factor Erythroid 2-Related Factor 2 (Nrf2) and levels of phosphorylated Nrf2, Heme Oxygenase 1 (HO-1), and Superoxide Dismutase (SOD) (p < 0.01). CONCLUSION TYP can regulate inflammation and oxidative stress in RA-FLS cells by activating the Keap1/Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Lu Tang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Mingquan Li
- Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Songlan Piao
- Clinical Medical School, Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Lianyun Du
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Saiyue Qiu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xin Jiang
- College of Integrative Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Meixiu Luo
- College of Integrative Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yinghang Wang
- The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Zhi Pan
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
41
|
Van Acker ZP, Leroy T, Annaert W. Mitochondrial dysfunction, cause or consequence in neurodegenerative diseases? Bioessays 2025; 47:e2400023. [PMID: 39367555 DOI: 10.1002/bies.202400023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 07/29/2024] [Accepted: 09/20/2024] [Indexed: 10/06/2024]
Abstract
Neurodegenerative diseases encompass a spectrum of conditions characterized by the gradual deterioration of neurons in the central and peripheral nervous system. While their origins are multifaceted, emerging data underscore the pivotal role of impaired mitochondrial functions and endolysosomal homeostasis to the onset and progression of pathology. This article explores whether mitochondrial dysfunctions act as causal factors or are intricately linked to the decline in endolysosomal function. As research delves deeper into the genetics of neurodegenerative diseases, an increasing number of risk loci and genes associated with the regulation of endolysosomal and autophagy functions are being identified, arguing for a downstream impact on mitochondrial health. Our hypothesis centers on the notion that disturbances in endolysosomal processes may propagate to other organelles, including mitochondria, through disrupted inter-organellar communication. We discuss these views in the context of major neurodegenerative diseases including Alzheimer's and Parkinson's diseases, and their relevance to potential therapeutic avenues.
Collapse
Affiliation(s)
- Zoë P Van Acker
- Laboratory for Membrane Trafficking, VIB Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Thomas Leroy
- Laboratory for Membrane Trafficking, VIB Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Wim Annaert
- Laboratory for Membrane Trafficking, VIB Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
42
|
Guan XY, Dong X, Wang YX, Xu BC, Wu XB. Mitochondrial dysfunction in trigeminal ganglion contributes to nociceptive behavior in a nitroglycerin-induced migraine mouse model. Mol Pain 2025; 21:17448069251332100. [PMID: 40110756 PMCID: PMC12035203 DOI: 10.1177/17448069251332100] [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: 12/18/2024] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
Migraine is a chronic episodic neurological disorder. However, its diagnosis and management remain unclear. The pathogenesis of migraine is intricately linked to the dysfunction of mitochondria and aberrant trigeminal neuronal activity. Here, we established a murine migraine model via intraperitoneal administration of nitroglycerin (NTG) to examine alterations in mitochondria-associated proteins and calcium signaling patterns within trigeminal neurons, while also investigating the underlying mechanisms. NTG-treated mice exhibited marked periorbital allodynia, decreased crossing of the central area, and decreased time spent in the central area in the open field test compared to Veh treated animals. Furthermore, increased calcium signaling in response to adenosine triphosphate (ATP) stimulation was observed in the trigeminal ganglion (TG) of mice with migraine. Meanwhile, mRNA levels of genes including nuclear respiratory factor-1 (Nrf1), nuclear respiratory factor-2 (Nrf2) and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (Pgc-1) were decreased in the TG. Pharmacological regulation of the mitochondrial function affected NTG-induced migraine chronic pain symptoms. TG mitochondria dysfunctions is implicated in the regulation of mechanical hyperalgesia through the modulation of calcium signaling in an NTG-induced migraine animal model.
Collapse
Affiliation(s)
- Xin-Ying Guan
- Department of Neurology, the Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Lianyungang, Jiangsu, China
| | - Xin Dong
- Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yi-Xuan Wang
- Department of Neurology, the Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Lianyungang, Jiangsu, China
| | - Bing-Chao Xu
- Department of Neurology, the Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Lianyungang, Jiangsu, China
| | - Xiao-Bo Wu
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
43
|
Olasehinde TA, Olaniran AO. Assessment of Neurotoxic Mechanisms of Individual and Binary Mixtures of Cobalt, Nickel and Lead in Hippocampal Neuronal Cells. ENVIRONMENTAL TOXICOLOGY 2025; 40:128-139. [PMID: 39365032 PMCID: PMC11628647 DOI: 10.1002/tox.24418] [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: 06/17/2023] [Revised: 06/14/2024] [Accepted: 08/20/2024] [Indexed: 10/05/2024]
Abstract
Many studies have focused on the neurotoxic effects of single metals, while investigation on the exposure to metal mixtures, which mainly occur in real-life situations, is scarce. This study sought to assess the neurotoxic effect of Ni, Co, and Pb binary mixtures and their individual effects in hippocampal neuronal cells (HT-22). Cells were exposed to Ni, Co, and Pb separately for 48 h at 37°C and 5% CO2, and cell viability was assessed. Morphological assessment of the cells exposed to binary mixtures of Co, Ni, and Pb and single metals was assessed using a microscope. Furthermore, acetylcholinesterase (AChE) activity, oxidative stress biomarkers (glutathione [GSH] and malondialdehyde [MDA] levels, catalase [CAT], and glutathione-S transferase [GST] activities) and nitric oxide [NO] levels were evaluated after treatment with the binary mixtures and single metals. Binary mixtures of the metals reduced cell viability, exerting an additivity action. The combinations also exerted synergistic action, as revealed by the combination index. Furthermore, a significant reduction in AChE activity, GSH levels, CAT and GST activities, and high MDA and NO levels were observed in neuronal cells. The additive interactions and synergistic actions of the binary mixtures might contribute to the significant reduction of AChE activity, GSH levels, GST, and CAT activities, and an increase in MDA and NO levels. The findings from this study revealed significant evidence that binary mixtures of Co, Pb, and Ni may induce impaired neuronal function and, ultimately, neurodegeneration.
Collapse
Affiliation(s)
- Tosin A. Olasehinde
- Nutrition and Toxicology Division, Food Technology DepartmentFederal Institute of Industrial ResearchLagosNigeria
- Discipline of Microbiology, School of Life SciencesUniversity of Kwazulu‐NatalDurbanSouth Africa
| | - Ademola O. Olaniran
- Discipline of Microbiology, School of Life SciencesUniversity of Kwazulu‐NatalDurbanSouth Africa
| |
Collapse
|
44
|
Agarwal U, Pannu A, Tonk RK. Foreign Contaminants Target Brain Health. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2025; 24:353-374. [PMID: 39812065 DOI: 10.2174/0118715273338071241213101016] [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: 06/17/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 01/16/2025]
Abstract
Neurodisease, caused by undesired substances, can lead to mental health conditions like depression, anxiety and neurocognitive problems like dementia. These substances can be referred to as contaminants that can cause damage, corruption, and infection or reduce brain functionality. Contaminants, whether conceptual or physical, have the ability to disrupt many processes. These observations motivate us to investigate contaminants and neurotoxicity collaboratively. This study investigates the link between pollutants and neuro-disease, examining transmission pathways and categorization. It also provides information on resources, causes, and challenges to minimize contamination risks. Contamination may cause various neuro-diseases, including Alzheimer's, Parkinson's, multi-system atrophy, Huntington's, autism spectrum disorder, psychiatric disorder, dementia, meningitis, encephalitis, schizophrenia, anxiety, and depression. The negative effects depend on the nature and extent of exposure. A comprehensive literature search was conducted using databases such as PubMed and Scopus, focusing on studies published till 2024. Studies were selected based on their examination of the relationship between environmental contaminants and brain health, emphasizing transmission pathways and the resulting neurological outcomes. Findings indicate that contaminants can penetrate the blood-brain barrier (BBB) via nasal, gut, and auditory routes, triggering harmful neurophysiological processes. This review highlights the urgent need for increased global awareness, policy interventions, and preventive measures to mitigate the long-term impacts of environmental contaminants on brain health, particularly in emerging nations.
Collapse
Affiliation(s)
- Uma Agarwal
- Department of Pharmaceutical Chemistry, Delhi Pharmaceutical Sciences & Research University, Delhi, 110017, India
| | - Arzoo Pannu
- Department of Pharmacology, Delhi Pharmaceutical Sciences & Research University, Delhi, 110017, India
| | - Rajiv Kumar Tonk
- Department of Pharmaceutical Chemistry, Delhi Pharmaceutical Sciences & Research University, Delhi, 110017, India
| |
Collapse
|
45
|
Guevara-Ramírez P, Tamayo-Trujillo R, Cadena-Ullauri S, Ruiz-Pozo V, Paz-Cruz E, Annunziata G, Verde L, Frias-Toral E, Simancas-Racines D, Zambrano AK. Heavy metals in the diet: unraveling the molecular pathways linked to neurodegenerative disease risk. FOOD AGR IMMUNOL 2024; 35. [DOI: 10.1080/09540105.2024.2434457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/21/2024] [Indexed: 01/03/2025] Open
Affiliation(s)
- Patricia Guevara-Ramírez
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Rafael Tamayo-Trujillo
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Santiago Cadena-Ullauri
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Viviana Ruiz-Pozo
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Elius Paz-Cruz
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Giuseppe Annunziata
- Facoltà di Scienze Umane, della Formazione e dello Sport, Università Telematica Pegaso, Naples, Italy
| | - Ludovica Verde
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | | | - Daniel Simancas-Racines
- Centro de Investigación de Salud Pública y Epidemiología Clínica (CISPEC), Universidad UTE, Quito, Ecuador
| | - Ana Karina Zambrano
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| |
Collapse
|
46
|
Mohd Murshid N, Mohd Sahardi NFN, Makpol S. Advancing Alzheimer's Disease Modelling by Developing a Refined Biomimetic Brain Microenvironment for Facilitating High-Throughput Screening of Pharmacological Treatment Strategies. Int J Mol Sci 2024; 26:241. [PMID: 39796097 PMCID: PMC11719782 DOI: 10.3390/ijms26010241] [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: 10/18/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 01/13/2025] Open
Abstract
Alzheimer's disease (AD) poses a significant worldwide health challenge, requiring novel approaches for improved models and treatment development. This comprehensive review emphasises the systematic development and improvement of a biomimetic brain environment to address the shortcomings of existing AD models and enhance the efficiency of screening potential drug treatments. We identify drawbacks in traditional models and emphasise the necessity for more physiologically accurate systems through an in-depth analysis of current literature. This review aims to study the development of an advanced AD model that accurately replicates key AD pathophysiological aspects using cutting-edge biomaterials and microenvironment design. Incorporating biomolecular elements like Tau proteins and beta-amyloid (Aβ) plaques improve the accuracy of illustrating disease mechanisms. The expected results involve creating a solid foundation for high-throughput screening with enhanced scalability, translational significance, and the possibility of speeding up drug discovery. Thus, this review fills the gaps in AD modelling and shows potential for creating precise and efficient drug treatments for AD.
Collapse
Affiliation(s)
- Nuraqila Mohd Murshid
- Department of Biochemistry, Faculty of Medicine, Level 17 Preclinical Building, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Nur Fatin Nabilah Mohd Sahardi
- Secretariat of Research and Innovation, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, Level 17 Preclinical Building, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia;
| |
Collapse
|
47
|
Zhuang X, Lin J, Song Y, Ban R, Zhao X, Xia Z, Wang Z, Zhang G. The Interplay Between Accumulation of Amyloid-Beta and Tau Proteins, PANoptosis, and Inflammation in Alzheimer's Disease. Neuromolecular Med 2024; 27:2. [PMID: 39751702 DOI: 10.1007/s12017-024-08815-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: 07/15/2024] [Accepted: 11/01/2024] [Indexed: 01/04/2025]
Abstract
Alzheimer's disease (AD) is a common progressive neurodegenerative disorder, and the vast majority of cases occur in elderly patients. Recently, the accumulation of Aβ and tau proteins has drawn considerable attention in AD research. This review explores the multifaceted interactions between these proteins and their contribution to the pathological landscape of AD, encompassing synaptic dysfunction, neuroinflammation, and PANoptosis. PANoptosis is a collective term for programmed cell death (PCD) modalities that encompass elements of apoptosis, pyroptosis, and necroptosis. The accumulation of Aβ peptides and tau proteins, along with the immune response in brain cells, may trigger PANoptosis, thus advancing the progression of the disease. Recent advancements in molecular imaging and genetics have provided deeper insights into the interactions between Aβ peptides, tau proteins, and the immune response. The review also discusses the role of mitochondrial dysregulation in AD. The exploration of the interplay between neurodegeneration, immune responses, and cell death offers promising avenues for the development of innovative treatments.
Collapse
Affiliation(s)
- Xianbo Zhuang
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, 252000, China
| | - Jie Lin
- School of Basic Medicine Sciences, Shandong University, Jinan, China
- Department of Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, China
| | - Yamin Song
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, 252000, China
| | - Ru Ban
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, 252000, China
| | - Xin Zhao
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, 252000, China
| | - Zhangyong Xia
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, 252000, China.
- Department of Neurology, Liaocheng People's Hospital, Shandong University, Jinan, 250012, China.
- Department of Neurology, the Second People's Hospital of Liaocheng, Liaocheng, China.
| | - Zheng Wang
- Department of Neurosurgery, Liaocheng Traditional Chinese Medicine Hospital, Liaocheng, 252000, China.
| | - Guifeng Zhang
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, 252000, China.
| |
Collapse
|
48
|
Montanari M, Mercuri NB, Martella G. Exceeding the Limits with Nutraceuticals: Looking Towards Parkinson's Disease and Frailty. Int J Mol Sci 2024; 26:122. [PMID: 39795979 PMCID: PMC11719863 DOI: 10.3390/ijms26010122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
One of the most pressing challenges facing society today is the rising prevalence of physical and cognitive frailty. This geriatric condition makes older adults more vulnerable to disability, illness, and a heightened risk of mortality. In this scenario, Parkinson's disease (PD) and geriatric frailty, which share several common characteristics, are becoming increasingly prevalent worldwide, underscoring the urgent need for innovative strategies. Nutraceuticals are naturally occurring bioactive compounds contained in foods, offering health benefits over and above essential nutrition. By examining the literature from the past decade, this review highlights how nutraceuticals can act as complementary therapies, addressing key processes, such as oxidative stress, inflammation, and neuroprotection. Notably, the antioxidant action of nutraceuticals appears particularly beneficial in regard to PD and geriatric frailty. For instance, antioxidant-rich nutraceuticals may mitigate the oxidative damage linked to levodopa therapy in PD, potentially reducing the side effects and enhancing treatment sustainability. Similarly, the antioxidant effects of nutraceuticals may amplify the benefits of physical activity, enhancing muscle function, cognitive health, and resilience, thereby reducing the risk of frailty. This review proposes a holistic approach integrating nutraceuticals with exercise, pharmacotherapy, and lifestyle adjustments. It promises to transform the management of ARD, prolong life, and improve the quality of life and well-being of older people.
Collapse
Affiliation(s)
- Martina Montanari
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy;
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
| | - Nicola Biagio Mercuri
- Neurology Unit, Policlinico Tor Vergata, University of Rome Tor Vergata, 00133 Rome, Italy;
- Department of Experimental Neuroscience, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
- Department of Wellbeing, Nutrition and Sport, Faculty of Humanities Educations and Sports, Pegaso Telematics University, 80145 Naples, Italy
| |
Collapse
|
49
|
Li J, Xu X, Yang S, Liu K, Wu M, Xie M, Xiong T. Helicobacter pylori Inhibition, Gastritis Attenuation, and Gut Microbiota Protection in C57BL/6 Mice by Ligilactobacillus salivarius NCUH062003. Microorganisms 2024; 12:2521. [PMID: 39770724 PMCID: PMC11678540 DOI: 10.3390/microorganisms12122521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Helicobacter pylori (H. pylori), one of the most prevalent pathogenic bacteria worldwide, is the leading cause of gastritis, gastric intestinal metaplasia, and gastric cancer. Antibiotics, the conventional treatment for eliminating H. pylori, often lead to severe bacterial resistance, gut dysbiosis, and hepatic insufficiency and fail to address the inflammatory response or gastric mucosal damage caused by H. pylori infection. In this study, based on 10-week animal experiments, two models of L. salivarius NCUH062003 for the prophylaxis and therapy of H. pylori infection in C57BL/6 mice were established; a comprehensive comparative analysis was performed to investigate the anti-H. pylori effect of probiotics, the reduction in inflammation, and repair of gastric mucosal damage. ELISA, immunohistochemistry, and pathology analyses showed that NCUH062003 decreased the expression of pro-inflammatory cytokine interleukins (IL-1β, IL-6) and myeloperoxidase (MPO) and reduced neutrophil infiltration in the gastric mucosa lamina propria. Immunofluorescence and biochemical analysis showed that NCUH062003 resisted gastric epithelial cell apoptosis, increased the level of superoxide dismutase (SOD) in gastric mucosa, and promoted the expression of tight junction protein ZO1 and Occludin. In addition, through high-throughput sequencing, in the probiotic therapy and prophylactic mode, the diversity and composition of the gut microbiota of HP-infected mice were clarified, the potential functions of the gut microbiota were analyzed, the levels of short-chain fatty acids (SCFAs) were measured, and the effects of L. salivarius NCUH062003 on the gut microbiota and its metabolites in HP-infected mice treated with amoxicillin/metronidazole were revealed. This study provides functional strain resources for the development and application of microbial agents seeking to antagonize H. pylori beyond antibiotics.
Collapse
Affiliation(s)
- Junyi Li
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China; (J.L.)
- School of Food Science & Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
| | - Xiaoyan Xu
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China; (J.L.)
- School of Food Science & Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
| | - Shiyu Yang
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China; (J.L.)
- School of Food Science & Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
| | - Kui Liu
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China; (J.L.)
- School of Food Science & Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
| | - Min Wu
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China; (J.L.)
- School of Food Science & Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
| | - Mingyong Xie
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China; (J.L.)
- School of Food Science & Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
| | - Tao Xiong
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China; (J.L.)
- School of Food Science & Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
| |
Collapse
|
50
|
Boziki M, Theotokis P, Kesidou E, Nella M, Bakirtzis C, Karafoulidou E, Tzitiridou-Chatzopoulou M, Doulberis M, Kazakos E, Deretzi G, Grigoriadis N, Kountouras J. Impact of Mast Cell Activation on Neurodegeneration: A Potential Role for Gut-Brain Axis and Helicobacter pylori Infection. Neurol Int 2024; 16:1750-1778. [PMID: 39728753 DOI: 10.3390/neurolint16060127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/08/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND The innate immune response aims to prevent pathogens from entering the organism and/or to facilitate pathogen clearance. Innate immune cells, such as macrophages, mast cells (MCs), natural killer cells and neutrophils, bear pattern recognition receptors and are thus able to recognize common molecular patterns, such as pathogen-associated molecular patterns (PAMPs), and damage-associated molecular patterns (DAMPs), the later occurring in the context of neuroinflammation. An inflammatory component in the pathology of otherwise "primary cerebrovascular and neurodegenerative" disease has recently been recognized and targeted as a means of therapeutic intervention. Activated MCs are multifunctional effector cells generated from hematopoietic stem cells that, together with dendritic cells, represent first-line immune defense mechanisms against pathogens and/or tissue destruction. METHODS This review aims to summarize evidence of MC implication in the pathogenesis of neurodegenerative diseases, namely, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, and multiple sclerosis. RESULTS In view of recent evidence that the gut-brain axis may be implicated in the pathogenesis of neurodegenerative diseases and the characterization of the neuroinflammatory component in the pathology of these diseases, this review also focuses on MCs as potential mediators in the gut-brain axis bi-directional communication and the possible role of Helicobacter pylori, a gastric pathogen known to alter the gut-brain axis homeostasis towards local and systemic pro-inflammatory responses. CONCLUSION As MCs and Helicobacter pylori infection may offer targets of intervention with potential therapeutic implications for neurodegenerative disease, more clinical and translational evidence is needed to elucidate this field.
Collapse
Affiliation(s)
- Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Maria Nella
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Christos Bakirtzis
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Eleni Karafoulidou
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Maria Tzitiridou-Chatzopoulou
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- Midwifery Department, School of Healthcare Sciences, University of West Macedonia, Koila, 50100 Kozani, Greece
| | - Michael Doulberis
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- Gastroklinik, Private Gastroenterological Practice, 8810 Horgen, Switzerland
- Division of Gastroenterology and Hepatology, Medical University Department, 5001 Aarau, Switzerland
| | - Evangelos Kazakos
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| | - Georgia Deretzi
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- Department of Neurology, Papageorgiou General Hospital, 54629 Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Jannis Kountouras
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| |
Collapse
|