1
|
Agadagba SK, Yau SY, Liang Y, Dalton K, Thompson B. Bidirectional causality of physical exercise in retinal neuroprotection. Neural Regen Res 2025; 20:3400-3415. [PMID: 39688575 PMCID: PMC11974656 DOI: 10.4103/nrr.nrr-d-24-00942] [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: 08/16/2024] [Revised: 10/21/2024] [Accepted: 11/16/2024] [Indexed: 12/18/2024] Open
Abstract
Physical exercise is recognized as an effective intervention to improve mood, physical performance, and general well-being. It achieves these benefits through cellular and molecular mechanisms that promote the release of neuroprotective factors. Interestingly, reduced levels of physical exercise have been implicated in several central nervous system diseases, including ocular disorders. Emerging evidence has suggested that physical exercise levels are significantly lower in individuals with ocular diseases such as glaucoma, age-related macular degeneration, retinitis pigmentosa, and diabetic retinopathy. Physical exercise may have a neuroprotective effect on the retina. Therefore, the association between reduced physical exercise and ocular diseases may involve a bidirectional causal relationship whereby visual impairment leads to reduced physical exercise and decreased exercise exacerbates the development of ocular disease. In this review, we summarize the evidence linking physical exercise to eye disease and identify potential mediators of physical exercise-induced retinal neuroprotection. Finally, we discuss future directions for preclinical and clinical research in exercise and eye health.
Collapse
Affiliation(s)
- Stephen K. Agadagba
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
| | - Suk-yu Yau
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Ying Liang
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
| | - Kristine Dalton
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, Canada
| | - Benjamin Thompson
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
2
|
Chen R, Chen W, Li P, Zhao Y, Zeng Q, Chen W, Cao D. Function and application of brain‑derived neurotrophic factor precursors (Review). Int J Mol Med 2025; 56:105. [PMID: 40341415 PMCID: PMC12081033 DOI: 10.3892/ijmm.2025.5546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 04/29/2025] [Indexed: 05/10/2025] Open
Abstract
Brain‑derived neurotrophic factor precursor (proBDNF) plays a critical role in the pathogenesis and progression of various human diseases. Through its interaction with p75NTR and sortilin receptors, proBDNF promotes apoptosis, impairs synaptic plasticity, and contributes to the regulation of immune system function, inflammatory responses and cellular metabolic processes. proBDNF is widely distributed throughout the body, and as such, extensive research has demonstrated that proBDNF is significantly associated with the pathophysiological mechanisms underlying several diseases. In the present review, the mechanisms by which proBDNF contributes to different diseases are summarized to highlight its potential therapeutic and diagnostic implications. Specifically, the role of proBDNF in cognitive disorders, focusing on its effects on synaptic function and neural network dynamics, while analyzing the cascade reactions involving proBDNF and downstream effector molecules in inflammatory diseases, to elucidate its bidirectional regulatory effects in tumor initiation and progression. Furthermore, the function of proBDNF in neurogenesis, the mechanism by which it regulates the memory of fear, and enhances individual behavioral flexibility is discussed. Finally, the potential of proBDNF as a biomarker for disease diagnosis and the therapeutic prospects of targeting it using monoclonal antibodies are highlighted while also proposing future research directions. The present review can serve as a reference for translational medical research on proBDNF and its receptors.
Collapse
Affiliation(s)
- Risheng Chen
- Department of Anesthesiology, Pingshan Central Hospital of Shenzhen, Shenzhen, Guangdong 518122, P.R. China
| | - Weixin Chen
- Department of Science and Education, The Fourth People's Hospital of Shenzhen (Shenzhen Sami Medical Center), Shenzhen, Guangdong 518118, P.R. China
| | - Ping Li
- Department of Anesthesiology, Pingshan Central Hospital of Shenzhen, Shenzhen, Guangdong 518122, P.R. China
| | - Yingchang Zhao
- Department of Anesthesiology, Pingshan Central Hospital of Shenzhen, Shenzhen, Guangdong 518122, P.R. China
| | - Qianqian Zeng
- Department of Anesthesiology, Pingshan Central Hospital of Shenzhen, Shenzhen, Guangdong 518122, P.R. China
| | - Wenqing Chen
- Department of Anesthesiology, Pingshan Central Hospital of Shenzhen, Shenzhen, Guangdong 518122, P.R. China
| | - Dequan Cao
- Department of Anesthesiology, Pingshan Central Hospital of Shenzhen, Shenzhen, Guangdong 518122, P.R. China
| |
Collapse
|
3
|
Orellana AM, Port's NMS, de Sá Lima L, Leite JA, Andreotti DZ, Kinoshita PF, Cantanzaro AB, Neto JAM, Scavone C, Kawamoto EM. Ouabain increases neuronal differentiation of hippocampal neural precursor cells. CURRENT RESEARCH IN NEUROBIOLOGY 2025; 8:100147. [PMID: 40166632 PMCID: PMC11957680 DOI: 10.1016/j.crneur.2025.100147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 04/02/2025] Open
Abstract
Image 1.
Collapse
Affiliation(s)
- Ana Maria Orellana
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Natacha Medeiros S. Port's
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Larissa de Sá Lima
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Jacqueline Alves Leite
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Goiás, 74045-155, Brazil
| | - Diana Zukas Andreotti
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Paula Fernanda Kinoshita
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Arthur B. Cantanzaro
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - João Agostinho M. Neto
- Laboratory of Cancer biology and Antineoplastic agents. Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Cristoforo Scavone
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| | - Elisa M. Kawamoto
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, Brazil
| |
Collapse
|
4
|
Kokkali M, Karali K, Thanou E, Papadopoulou MA, Zota I, Tsimpolis A, Efstathopoulos P, Calogeropoulou T, Li KW, Sidiropoulou K, Gravanis A, Charalampopoulos I. Multimodal beneficial effects of BNN27, a nerve growth factor synthetic mimetic, in the 5xFAD mouse model of Alzheimer's disease. Mol Psychiatry 2025; 30:2265-2283. [PMID: 39587294 PMCID: PMC12092300 DOI: 10.1038/s41380-024-02833-w] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/27/2024]
Abstract
Alzheimer's Disease (AD) is an incurable and debilitating progressive, neurodegenerative disorder which is the leading cause of dementia worldwide. Neuropathologically, AD is characterized by the accumulation of Aβ amyloid plaques in the microenvironment of brain cells and neurovascular walls, chronic neuroinflammation, resulting in neuronal and synaptic loss, myelin and axonal failure, as well as significant reduction in adult hippocampal neurogenesis. The hippocampal formation is particularly vulnerable to this degenerative process, due to early dysfunction of the cholinergic circuit. Neurotrophic factors consist major regulatory molecules and their decline in AD is considered as an important cause of disease onset and progression. Novel pharmacological approaches are targeting the downstream pathways controlled by neurotrophins, such as nerve growth factor (NGF) receptors, TrkA and p75NTR, which enhance hippocampal neurogenic capacity and neuroprotective mechanisms, and potentially counteract the neurotoxic effects of amyloid deposition. BNN27 is a non-toxic, newly developed 17-spiro-steroid analog, penetrating the blood-brain-barrier (BBB) and mimicking the neuroprotective effects of NGF, acting as selective activator of its receptors, both TrkA and p75NTR, thus promoting survival of various neuronal cell types. Our present research aims at determining whether and which aspects of the AD-related pathology, BNN27 is able to alleviate, exploring the cellular and molecular AD components and link these changes with improvements in the cognitive performance of an animal AD model, the 5xFAD mice. Our results clearly indicate that BNN27 administration significantly reduced amyloid-β load in whole brain of the animals, enhanced adult hippocampal neurogenesis, restored cholinergic function and synaptogenesis, reducing inflammatory activation and leading to significant restoration of cognitive functions. BNN27 may represent a new lead multimodal molecule with neuroprotective, neurogenic and anti-neuroinflammatory actions for developing druggable anti-Alzheimeric agents. Proteomics data are available via ProteomeXchange with the identifier PXD044699.
Collapse
Affiliation(s)
- Maria Kokkali
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece
| | - Kanelina Karali
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece
| | - Evangelia Thanou
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics & Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Maria Anna Papadopoulou
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece
| | - Ioanna Zota
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece
| | - Alexandros Tsimpolis
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece
| | | | | | - Ka Wan Li
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics & Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Kyriaki Sidiropoulou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece
- Department of Biology, School of Sciences and Engineering, University of Crete, Heraklion, 71003, Greece
| | - Achille Gravanis
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece
| | - Ioannis Charalampopoulos
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003, Greece.
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece.
| |
Collapse
|
5
|
Gaesser GA, Hall SE, Angadi SS, Poole DC, Racette SB. Increasing the health span: unique role for exercise. J Appl Physiol (1985) 2025; 138:1285-1308. [PMID: 40244910 DOI: 10.1152/japplphysiol.00049.2025] [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: 01/23/2025] [Revised: 02/21/2025] [Accepted: 04/02/2025] [Indexed: 04/19/2025] Open
Abstract
Health span, that period between birth and onset of major disease(s), when adequate physical and cognitive function permit those daily living activities essential to life quality, is lower in the United States than other developed countries. Physical inactivity and excessive calorie intake occupy dominant roles both in the problem, and by redressing them, in the solution. Consequently, this review focuses on evidence that appropriate exercise engagement and calorie restriction (CR) can improve physical and mental health with a view to extending the health span. Humanity, writ large, has grasped these underlying concepts for Millennia but has been largely intransigent to them. Thus, the final section proposes a novel Monty Python-esque approach that encompasses humanity's inimical sense of humor to increase physical fitness and mental health, restore energy balance, sustain better cognitive function, and extend the health span.
Collapse
Affiliation(s)
- Glenn A Gaesser
- College of Health Solutions, Arizona State University, Phoenix, Arizona, United States
| | - Stephanie E Hall
- Colleges of Veterinary Medicine and Health and Human Sciences, Kansas State University, Manhattan, Kansas, United States
| | - Siddhartha S Angadi
- School of Health Education and Human Development, University of Virginia, Charlottesville, Virginia, United States
| | - David C Poole
- Colleges of Veterinary Medicine and Health and Human Sciences, Kansas State University, Manhattan, Kansas, United States
| | - Susan B Racette
- College of Health Solutions, Arizona State University, Phoenix, Arizona, United States
| |
Collapse
|
6
|
Athanasiou C, Claveras Cabezudo A, Tsengenes A, Wade RC. Simulation of Neurotrophin Receptor Transmembrane Helix Interactions Reveals Active States and Distinct Signaling Mechanisms. JACS AU 2025; 5:2186-2200. [PMID: 40443888 PMCID: PMC12117394 DOI: 10.1021/jacsau.5c00174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 06/02/2025]
Abstract
Neurotrophin (NT) receptor signaling regulates neuronal survival, axonal and dendritic network maintenance, differentiation, and synaptic plasticity. Signaling is initiated by binding of NT to the extracellular domain of NT receptor dimers, leading to activation of the receptor and signal propagation intracellularly. How this activating signal is mediated by the single-pass transmembrane (TM) helical domain of the receptor and what the relation between domain sequence and signaling mechanism is remain unclear. The structure and dynamics of the TM domain of the receptor dimers in the active and inactive states for intracellular signaling are still elusive, with NMR structures capturing only a single state. Here, we carried out unbiased and enhanced sampling molecular dynamics simulations of the TM domain dimers of the wild-type p75, TrkA and TrkB NT receptors and selected mutants in micelle and bilayer lipid environments at atomistic and coarse-grained levels of representation. The coarse-grained simulations enabled exploration of multiple states of the TM domain dimers and revealed the influence of the lipid environment on the TM helix arrangements. From the simulations, we identify active and inactive TM helix arrangements of the p75 and TrkA receptors that are supported by experimental data and suggest two different signaling mechanisms through the C-terminal regions of the TM helices. For TrkB, a single dominant but less energetically stable arrangement of the TM domain dimer is observed. These findings have implications for mechanistic studies of NT receptor signaling and the design of neuroprotective drugs to stabilize specific states of the TM domain of the receptors.
Collapse
Affiliation(s)
- Christina Athanasiou
- Molecular
and Cellular Modeling Group, Heidelberg
Institute for Theoretical Studies (HITS), 69118Heidelberg, Germany
- Faculty
of Biosciences, Heidelberg University, 69120Heidelberg, Germany
- Heidelberg
Biosciences International Graduate School, Heidelberg University, 69120Heidelberg, Germany
| | - Ainara Claveras Cabezudo
- Molecular
and Cellular Modeling Group, Heidelberg
Institute for Theoretical Studies (HITS), 69118Heidelberg, Germany
- Faculty
of Biosciences, Heidelberg University, 69120Heidelberg, Germany
| | - Alexandros Tsengenes
- Molecular
and Cellular Modeling Group, Heidelberg
Institute for Theoretical Studies (HITS), 69118Heidelberg, Germany
- Faculty
of Biosciences, Heidelberg University, 69120Heidelberg, Germany
- Heidelberg
Biosciences International Graduate School, Heidelberg University, 69120Heidelberg, Germany
| | - Rebecca C. Wade
- Molecular
and Cellular Modeling Group, Heidelberg
Institute for Theoretical Studies (HITS), 69118Heidelberg, Germany
- Faculty
of Biosciences, Heidelberg University, 69120Heidelberg, Germany
- Center
for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, 69120Heidelberg, Germany
- Interdisciplinary
Center for Scientific Computing (IWR), Heidelberg
University, 69120Heidelberg, Germany
| |
Collapse
|
7
|
Brunner SM, Gaisbauer S, Pallier PN, Yip PK, Ramspacher A, Leitner J, Sternberg F, Erhardt-Kreutzer C, Haslauer T, Huber S, Bieler L, Couillard-Despres S, Kofler B. Impact of galanin receptors 2 and 3 double-knockout on neuroinflammation and functional recovery following traumatic brain injury. Peptides 2025:171415. [PMID: 40412555 DOI: 10.1016/j.peptides.2025.171415] [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: 03/10/2025] [Revised: 05/16/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
Traumatic brain injury (TBI) is one of the world's leading causes of death and disability in young individuals and the mechanism underlying TBI-associated neuroinflammation is poorly understood. The regulatory neuropeptide galanin (GAL) and its three receptors (GAL1-3R) are assumed to modulate the neuroinflammatory response following TBI, especially by signalling via GAL2R and GAL3R. Therefore, the role of GALRs in acute neuroinflammation and functional recovery following moderate Controlled Cortical Impact TBI was studied using GAL2/3R-double-KO (GAL2/3R-KO) mice. Brains and cerebrospinal fluid (CSF) were collected at day 1 and 30 days post TBI. Functional recovery post TBI was assessed by the modified Neurological Severity Score (mNSS), Elevated Plus Maze (EPM) and Morris Water Maze (MWM) test. Post TBI (day 1 to 28 post injury), neurological dysfunction was more severe in GAL2/3R-KO mice than in WT mice. At 1 day post TBI, inflammatory markers and several nerve growth factors significantly increased in the ipsilateral hemisphere, compared to the contralateral hemisphere in both GAL2/3R-KO and WT mice. At 4 days post surgery, TBI mice entered significantly more frequent the open-arms in the EPM compared to Sham-operated mice, suggestive of increased exploratory behaviour in TBI mice. At 30 days post TBI, immunostaining of brain sections revealed significant differences in vascularisation and glial scarring in the cortex when comparing TBI and Sham-operated mice, but genotypes were similar. In summary, the results indicate that GAL2R and/or GAL3R have a neuroprotective role following moderate TBI, as the severity was significantly lower in their presence than in their absence.
Collapse
Affiliation(s)
- Susanne M Brunner
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria.
| | - Stefanie Gaisbauer
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria.
| | - Patrick N Pallier
- Centre for Neuroscience, Surgery and Trauma, The Blizard Institute, Queen Mary University of London, United Kingdom.
| | - Ping K Yip
- Centre for Neuroscience, Surgery and Trauma, The Blizard Institute, Queen Mary University of London, United Kingdom.
| | - Andrea Ramspacher
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria.
| | - Julia Leitner
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria.
| | - Felix Sternberg
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria.
| | - Christina Erhardt-Kreutzer
- Institute of Experimental Neurogeneration, Paracelsus Medical University, Salzburg, Austria; Department of General, Visceral and Thoracic Surgery, University Hospital of the Paracelsus Medical University, Salzburg, Austria.
| | - Theresa Haslauer
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria.
| | - Sara Huber
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria.
| | - Lara Bieler
- Institute of Experimental Neurogeneration, Paracelsus Medical University, Salzburg, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| | - Sebastien Couillard-Despres
- Institute of Experimental Neurogeneration, Paracelsus Medical University, Salzburg, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
8
|
Mitrovic M, Selakovic D, Jovicic N, Ljujic B, Rosic G. BDNF/proBDNF Interplay in the Mediation of Neuronal Apoptotic Mechanisms in Neurodegenerative Diseases. Int J Mol Sci 2025; 26:4926. [PMID: 40430064 PMCID: PMC12112594 DOI: 10.3390/ijms26104926] [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: 04/19/2025] [Revised: 05/12/2025] [Accepted: 05/20/2025] [Indexed: 05/29/2025] Open
Abstract
The neurotrophic system includes neurotrophins, such as brain-derived neurotrophic factor (BDNF) and its precursor proBDNF, which play conflicting roles in neuronal survival and apoptosis, with their balance having a significant impact on neurodegenerative outcomes. While BDNF is widely acknowledged as a potent neurotrophin that promotes neuronal survival and differentiation, its precursor, proBDNF, has the opposite effect, promoting apoptosis and neuronal death. This review highlights the new and unique aspects of BDNF/proBDNF interaction in the modulation of neuronal apoptotic pathways in neurodegenerative disorders. It systematically discusses the cross-talk in apoptotic signaling at the molecular level, whereby BDNF activates survival pathways such as PI3K/Akt and MAPK/ERK, whereas proBDNF activates p75NTR and sortilin to induce neuronal apoptosis via JNK, RhoA, NFkB, and Rac-GTPase pathways such as caspase activation and mitochondrial injury. Moreover, this review emphasizes the factors that affect the balance between proBDNF and BDNF levels within the context of neurodegeneration, including proteolytic processing, the expression of TrkB and p75NTR receptors, and extrinsic gene transcription regulators. Cellular injury, stress, or signaling pathway alterations can disrupt the balance of BDNF/proBDNF, which may be involved in apoptotic-related neurodegenerative diseases like Alzheimer's, Parkinson's, and Huntington's diseases. This review provides a comprehensive framework for targeting neurotrophin signaling in the development of innovative therapies for neuronal survival and managing apoptotic-related neurodegenerative disorders, addressing the mechanistic complexity and clinical feasibility of BDNF/proBDNF interaction.
Collapse
Affiliation(s)
- Marina Mitrovic
- Department of Medical Biochemistry, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Dragica Selakovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Nemanja Jovicic
- Department of Histology and Embryology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Biljana Ljujic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Gvozden Rosic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| |
Collapse
|
9
|
Han H, Yao J, Wu J, Mao S, Pan H, Qv L, Zhu G, Ren J, Yu Y, Xuan F, Zeng L, Ma Y, Yang Z, Zhu Z, Zhu F, Li MD. Implications of neurogenesis in depression through BDNF: rodent models, regulatory pathways, gut microbiota, and potential therapy. Mol Psychiatry 2025:10.1038/s41380-025-03044-7. [PMID: 40341897 DOI: 10.1038/s41380-025-03044-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 03/05/2025] [Accepted: 04/30/2025] [Indexed: 05/11/2025]
Abstract
Major Depressive Disorder (MDD) is a prevalent psychiatric disorder with a profound impact on global health, necessitating a deeper understanding of its pathophysiology. This review synthesizes current evidence linking neurogenesis, particularly in the hippocampal region, with MDD. Accumulating data showed a significant reduction of neurogenesis in the hippocampal region of both MDD patients and various MDD rodent models. We highlight the role of brain-derived neurotrophic factor (BDNF) and its associated signaling pathways in regulating neurogenesis and depressive symptoms. Additionally, the influence of gut microbiota on the neurogenesis in depression is presented, offering a novel perspective on environmental modulation of neurogenesis. This review also underscores the potential antidepressant interventions targeting neurogenesis and BDNF's regulation, such as therapeutic benefits of environmental enrichment, physical activity, and pharmacological agents in enhancing neurogenesis and alleviating depressive symptoms. Together, this systemic review provides a foundation for future research aiming at developing personalized treatments by targeting neurogenesis in MDD, potentially leading to novel biomarkers and therapeutic strategies.
Collapse
Affiliation(s)
- Haijun Han
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Jianhua Yao
- Joint Institute of Tobacco and Health, Kunming, Yunnan, China
| | - Jinhan Wu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Shiqi Mao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Hongyi Pan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Lingling Qv
- Central Laboratory, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Guanqi Zhu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Juntian Ren
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Yaning Yu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Feiyang Xuan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Linghui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Yunlong Ma
- Department of Psychiatry, Perelman School of Medicine, Lifespan Brain Institute at Penn Med and the Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhongli Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhijing Zhu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Feng Zhu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of Brain and Cognitive Science, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Ming D Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
10
|
Wu J, Tang J, Huang D, Wang Y, Zhou E, Ru Q, Xu G, Chen L, Wu Y. Study on the comorbid mechanisms of sarcopenia and late-life depression. Behav Brain Res 2025; 485:115538. [PMID: 40122287 DOI: 10.1016/j.bbr.2025.115538] [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] [Revised: 03/06/2025] [Accepted: 03/09/2025] [Indexed: 03/25/2025]
Abstract
The increasing global aging population has brought greater focus to age-related diseases, particularly muscle-brain comorbidities such as sarcopenia and late-life depression. Sarcopenia, defined by the gradual loss of muscle mass and function, is notably prevalent among older individuals, while late-life depression profoundly affects their mental health and overall well-being. Epidemiological evidence suggests a high co-occurrence of these two conditions, although the precise biological mechanisms linking them remain inadequately understood. This review synthesizes the existing body of literature on sarcopenia and late-life depression, examining their definitions, prevalence, clinical presentations, and available treatments. The goal is to clarify the potential connections between these comorbidities and offer a theoretical framework for the development of future preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Jiale Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Jun Tang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Di Huang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Yu Wang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Enyuan Zhou
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Guodong Xu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| |
Collapse
|
11
|
Won CW, Kim M, Shin HE. From a Solitary Blood-Derived Biomarker to Combined Biomarkers of Sarcopenia: Experiences From the Korean Frailty and Aging Cohort Study. J Gerontol A Biol Sci Med Sci 2025; 80:glae237. [PMID: 39417263 DOI: 10.1093/gerona/glae237] [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: 02/15/2024] [Indexed: 10/19/2024] Open
Abstract
Sarcopenia is recognized as a complex and multifactorial disorder that includes nutritional deficiency, inactivity, proinflammatory status, hormonal changes, neurological degeneration, and metabolic disturbances. It's pathogenesis is not fully understood. Therefore, identifying specific biomarkers of sarcopenia will help us understand its pathophysiology. The most frequently reported blood-derived biomarkers of sarcopenia are growth factors, neuromuscular junctions, endocrine systems, mitochondrial dysfunction, inflammation-mediated and redox processes, muscle protein turnover, blood metabolomics, and behavior-mediated biomarkers. Here, we address the implications of sarcopenia biomarkers based on our research experience with Korean Frailty and Aging Cohort Study cohort data. It includes free testosterone, myostatin, fibroblast growth factor 21 (FGF-21), growth differentiation factor 15 (GDF-15), procollagen type III N-terminal peptide (P3NP), creatinine-based biomarkers, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), brain-derived neurotrophic factor (BDNF), metabolites (proline, alanine, tryptophan), and multi-biomarker risk score. We attempted to explain the paradoxical findings of myostatin and FGF-21 levels in relation to sarcopenia. GDF-15 levels were associated with sarcopenia prevalence but not its incidence. Plasma P3NP and BDNF levels may be biomarkers of muscle quality rather than quantity. Lower erythrocyte eicosapentaenoic acid (EPA) and docosahexaenoic acid levels were associated with slow gait speed, and erythrocyte EPA levels were associated with low handgrip strength. We developed a multi-biomarker risk score for sarcopenia and found that its accuracy in diagnosing sarcopenia was higher than that of any single biomarker.
Collapse
Affiliation(s)
- Chang Won Won
- Department of Family Medicine, Elderly Frailty Sarcopenia Research Center, College of Medicine, Kyung Hee University, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Miji Kim
- Department of Health Sciences and Technology, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyung Eun Shin
- Department of Health Sciences and Technology, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
- Department of Orthopaedics, Emory Musculoskeletal Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
12
|
Kelliny S, Zhou X, Bobrovskaya L. Alzheimer's Disease and Frontotemporal Dementia: A Review of Pathophysiology and Therapeutic Approaches. J Neurosci Res 2025; 103:e70046. [PMID: 40387258 PMCID: PMC12087441 DOI: 10.1002/jnr.70046] [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: 12/19/2024] [Revised: 04/01/2025] [Accepted: 05/02/2025] [Indexed: 05/20/2025]
Abstract
Alzheimer's disease (AD) is a devastating form of dementia, with the number of affected individuals rising sharply. The main hallmarks of the disease include amyloid-beta plaque deposits and neurofibrillary tangles consisting of hyperphosphorylated tau protein, besides other pathological features that contribute to the disease's complexity. The causes of sporadic AD are multifactorial and mostly age-related and involve risk factors such as diabetes and cardiovascular or cerebrovascular disorders. Frontotemporal dementia (FTD) is another type of dementia characterized by a spectrum of behaviors, memory, and motor abnormalities and associated with abnormal depositions of protein aggregation, including tau protein. Currently approved medications are symptomatic, and no disease-modifying therapy is available to halt the disease progression. Therefore, the development of multi-targeted therapeutic approaches could hold promise for the treatment of AD and other neurodegenerative disorders, including tauopathies. In this article, we will discuss the pathophysiology of AD and FTD, the proposed hypotheses, and current therapeutic approaches, highlighting the development of novel drug candidates and the progress of clinical trials in this field of research.
Collapse
Affiliation(s)
- Sally Kelliny
- Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
- Faculty of PharmacyAssiut UniversityAssiutEgypt
| | - Xin‐Fu Zhou
- Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Larisa Bobrovskaya
- Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| |
Collapse
|
13
|
Wood LB, Singer AC. Neurons as Immunomodulators: From Rapid Neural Activity to Prolonged Regulation of Cytokines and Microglia. Annu Rev Biomed Eng 2025; 27:55-72. [PMID: 39805040 DOI: 10.1146/annurev-bioeng-110122-120158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Regulation of the brain's neuroimmune system is central to development, normal function, and disease. Neuronal communication to microglia, the primary immune cells of the brain, is well known to involve purinergic signaling mediated via ATP secretion and the cytokine fractalkine. Recent evidence shows that neurons release multiple cytokines beyond fractalkine, yet these are less studied and poorly understood. In contrast to ATP, cytokines are a class of signaling molecule that are much larger, with longer signaling and farther diffusion. We posit that neuron-expressed cytokines are an essential mechanism of neuron-microglia communication that arises as part of both normal learning and memory and in response to tissue pathology. Thus, neurons are underappreciated immunomodulatory cells that express diverse immunomodulatory signals. While neuronally sourced cytokines have been understudied, new technical advances make this a timely topic. The goal of this review is to define what is known about the cytokines expressed from neurons, how they are regulated, and the effects of these cytokines on microglia. We delineate key knowledge gaps and needs for new tools to define and analyze neuronal roles in immunomodulation. Given that cytokines are central regulators of microglial function, a broad new body of work is required to illuminate functional links between these neuronally expressed cytokines and sustained and transient microglial function.
Collapse
Affiliation(s)
- Levi B Wood
- Wallace H. Coulter Department of Biomedical Engineering, George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA; ,
| | - Annabelle C Singer
- Wallace H. Coulter Department of Biomedical Engineering, George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA; ,
| |
Collapse
|
14
|
Cui K, Zhou Y, Zhang L, Ying Y, Xue Y, Zhang X, Wang Q, Shen H, Zhou W, Gao F, Wang Z. Light Treatment Ameliorates Sub-chronic MK-801-Induced Cognitive Deficits in Mice Through Up-regulating BDNF/p-CREB/p-ERK Signaling Pathway. Mol Neurobiol 2025; 62:5947-5960. [PMID: 39673660 DOI: 10.1007/s12035-024-04653-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: 03/22/2023] [Accepted: 11/27/2024] [Indexed: 12/16/2024]
Abstract
Cognitive impairment associated with schizophrenia (CIAS) is considered a core symptom of the illness, yet effective treatments remain limited. Light plays an important role in regulating cognitive functions. However, the potential of light treatment (LT) to improve CIAS remains unknown. The current study aimed to investigate the efficacy of LT on CIAS and explore the underlying molecular mechanisms in a CIAS animal model. The CIAS group and the control group were sub-chronically administered MK-801 and saline, respectively. Concurrently, the LT/CIAS group, consisting of CIAS mice, received LT exposure (3000 Lux, 2 h/day, for 3 weeks). Results showed a significant enhancement in cognitive performance among LT/CIAS mice, as evidenced by improvements in the novel object recognition (NOR) test, novel location recognition (NLR) test, and Morris water maze (MWM) compared to the CIAS group. Remarkably, these beneficial effects of LT persisted for over 4 weeks after the termination of LT. Furthermore, Golgi-cox staining unveiled an increased dendritic spine density and enhanced morphological complexity in hippocampal CA1 pyramidal neurons following 3 weeks of LT. Subsequent investigations revealed elevated levels of brain-derived neurotrophic factor (BDNF) and heightened phosphorylation of cAMP response element-binding phosphorylation protein (p-CREB) in the hippocampus of the LT/CIAS group compared to the CIAS group. Moreover, LT elevated the phosphorylated extracellular signal-regulated kinase (p-ERK) in the hippocampus of the LT/CIAS group relative to the CIAS group. In conclusion, the current study demonstrates that long-term LT effectively ameliorated sub-chronic MK-801-induced cognitive deficits in mice, and the altered dendritic spine density and morphology of CA1 pyramidal neurons were rescued in the LT/CIAS group, potentially through the up-regulation of the BDNF/p-CREB/p-ERK signaling pathway in LT/CIAS mice.
Collapse
Affiliation(s)
- Keke Cui
- Zhejiang Key Laboratory of Pathophysiology, Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Rd, Ningbo, 315211, Zhejiang, China
| | - Yiying Zhou
- Key Laboratory of Addiction Research of Zhejiang Province, Kang Ning Hospital, Ningbo, 315010, China
| | - Lizhi Zhang
- Zhejiang Key Laboratory of Pathophysiology, Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Rd, Ningbo, 315211, Zhejiang, China
| | - Yudong Ying
- Zhejiang Key Laboratory of Pathophysiology, Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Rd, Ningbo, 315211, Zhejiang, China
| | - Yan Xue
- Zhejiang Key Laboratory of Pathophysiology, Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Rd, Ningbo, 315211, Zhejiang, China
| | - Xiaoqin Zhang
- Key Laboratory of Addiction Research of Zhejiang Province, Kang Ning Hospital, Ningbo, 315010, China
- Zhejiang Key Laboratory of Pathophysiology, Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Rd, Ningbo, 315211, Zhejiang, China
| | - Qinwen Wang
- Zhejiang Key Laboratory of Pathophysiology, Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Rd, Ningbo, 315211, Zhejiang, China
| | - Haowei Shen
- Key Laboratory of Addiction Research of Zhejiang Province, Kang Ning Hospital, Ningbo, 315010, China
- Zhejiang Key Laboratory of Pathophysiology, Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Rd, Ningbo, 315211, Zhejiang, China
| | - Wenhua Zhou
- Key Laboratory of Addiction Research of Zhejiang Province, Kang Ning Hospital, Ningbo, 315010, China
| | - Feng Gao
- The Affiliated People's Hospital of Ningbo University, Ningbo, 315100, China.
| | - Zhengchun Wang
- The Affiliated People's Hospital of Ningbo University, Ningbo, 315100, China.
- Key Laboratory of Addiction Research of Zhejiang Province, Kang Ning Hospital, Ningbo, 315010, China.
- Zhejiang Key Laboratory of Pathophysiology, Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Rd, Ningbo, 315211, Zhejiang, China.
| |
Collapse
|
15
|
La Loggia O, Antunes DF, Aubin-Horth N, Taborsky B. Social Complexity During Early Development has Long-Term Effects on Neuroplasticity in the Social Decision-Making Network. Mol Ecol 2025; 34:e17738. [PMID: 40116137 DOI: 10.1111/mec.17738] [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: 07/23/2024] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025]
Abstract
In social species, early social experience shapes the development of appropriate social behaviours during conspecific interactions referred to as social competence. However, the underlying neuronal mechanisms responsible for the acquisition of social competence are largely unknown. A key candidate to influence social competence is neuroplasticity, which functions to restructure neural networks in response to novel experiences or alterations of the environment. One important mediator of this restructuring is the neurotrophin BDNF, which is well conserved among vertebrates. We studied the highly social fish Neolamprologus pulcher, in which the impact of early social experience on social competence has been previously shown. We investigated experimentally how variation in the early social environment impacts markers of neuroplasticity by analysing the relative expression of the bdnf gene and its receptors p75NTR and TrkB across nodes of the social decision-making network. In fish raised in larger groups, bdnf and TrkB were upregulated in the anterior tuberal nucleus, compared to fish raised in smaller groups, while TrkB was downregulated and bdnf was upregulated in the lateral part of the dorsal telencephalon. In the preoptic area (POA), all three genes were upregulated in fish raised in large groups, suggesting that early social experiences might lead to changes of the neuronal connectivity in the POA. Our results highlight the importance of early social experience in programming the constitutive expression of neuroplasticity markers, suggesting that the effects of early social experience on social competence might be due to changes in neuroplasticity.
Collapse
Affiliation(s)
- Océane La Loggia
- Institute for Ecology and Evolution, Behavioural Ecology Division, University of Bern, Bern, Switzerland
| | - Diogo F Antunes
- Institute for Ecology and Evolution, Behavioural Ecology Division, University of Bern, Bern, Switzerland
| | - Nadia Aubin-Horth
- Département de Biologie and Institut de Biologie Intégrative et Des Systèmes, Université Laval, Quebec, Canada
| | - Barbara Taborsky
- Institute for Ecology and Evolution, Behavioural Ecology Division, University of Bern, Bern, Switzerland
| |
Collapse
|
16
|
Wang S, Tang Y, Su G, Zheng L, Zhuang Y, Chen B, Fan H, Zhao M, Wu J. Absorption, transport, blood-brain barrier penetration, and neuroprotection of walnut peptides LR and LPI. Food Res Int 2025; 209:116305. [PMID: 40253149 DOI: 10.1016/j.foodres.2025.116305] [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: 11/16/2024] [Revised: 02/10/2025] [Accepted: 03/14/2025] [Indexed: 04/21/2025]
Abstract
The prerequisite for neuroprotective peptides to exert physiological effect is that they can across intestinal epithelial barrier and blood-brain barrier (BBB). The study was aimed to investigate the absorption, transportation and BBB permeability of walnut neuroprotective peptides LR and LPI using Caco-2 cell monolayer and in vivo imaging, and further to evaluate their underlying mechanisms through transcriptome sequencing analysis of zebrafish brain. Results showed that LR and LPI improved learning and memory impairment in scopolamine-induced zebrafish. Both peptides could be intactly transported in Caco-2 cells but via different mechanisms. LR was transported via both PepT1-mediated active route and tight junction-regulated passive paracellular route, while LPI was via PepT1 route only, with apparent permeability coefficient (30.18 ± 1.94) × 10-7 cm/s and (51.91 ± 3.49) × 10-7 cm/s, respectively. In vivo imaging of nude mice after FITC-labeling peptides administration further consolidated their ability of absorption, metabolic stability, and BBB penetration. Interestingly, LR exhibited better brain influx than that of LPI in nude mice. Additionally, transcriptome sequencing analysis demonstrated that LR and LPI improved learning and memory capacity by intervening cholinergic system, synaptic development and plasticity, neurotrophins, and oxidative stress, which were subsequent verified by biochemical measurement of zebrafish brain.
Collapse
Affiliation(s)
- Shuguang Wang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China; Food Science and Engineering, South China University of Technology, Guangzhou 510640, China; Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton T6G 2P5, Canada
| | - Yao Tang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| | - Guowan Su
- Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Lin Zheng
- Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Yongliang Zhuang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| | - Bifen Chen
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China; Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Hongbing Fan
- Department of Animal and Food Sciences, University of Kentucky, Lexington 40546, United States
| | - Mouming Zhao
- Food Science and Engineering, South China University of Technology, Guangzhou 510640, China.
| | - Jianping Wu
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton T6G 2P5, Canada
| |
Collapse
|
17
|
Chen S, Rao Y, Liu W, Yang Q, Wu G, Wu F, Gao Y, Li J, Fu J. Role of TRKC and NT-3 proteins in the progression and prognosis of colorectal cancer. Transl Cancer Res 2025; 14:2457-2469. [PMID: 40386265 PMCID: PMC12079258 DOI: 10.21037/tcr-2025-519] [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: 03/06/2025] [Accepted: 04/14/2025] [Indexed: 05/20/2025]
Abstract
Background Colorectal cancer (CRC) is a common tumor of the digestive system. In recent years, CRC has had the fourth highest incidence and second highest mortality rate among tumors worldwide. Therefore, it is important to identify new molecular markers, and examine their relationship with the clinicopathological features and prognosis of CRC patients. This study examined the correlation between the expression levels of the tropomyosin receptor kinase C (TRKC) and neurotrophic factor-3 (NT-3) proteins, and the clinicopathological features and prognosis of CRC patients. Methods In total, 141 paraffin-embedded specimens from patients who had undergone radical surgical resection for CRC were analyzed. All CRC diagnoses were confirmed by pathological examination. Corresponding adjacent normal tissues served as controls. Immunohistochemical staining was employed to assess the expression of the TRKC and NT-3 proteins in both the CRC and adjacent normal tissues. A subsequent statistical analysis was conducted to explore the associations between the expression levels of these proteins, and the clinicopathological features and prognostic outcomes of CRC patients. Results The expression level of TRKC was significantly higher in the CRC tissues than the adjacent normal tissues, while the expression level of NT-3 was significantly lower in the CRC tissues than the adjacent normal tissues, and the observed differences were statistically significant (P<0.001). Notably, TRKC expression was significantly correlated with the tumor site (P<0.05), lymph node metastasis (P<0.05), tumor node metastasis (TNM) stage (P<0.01), serum carcinoembryonic antigen (CEA) level (P<0.01), and serum carbohydrate antigen 19-9 (CA199) level (P<0.05). The patients with positive TRKC had significantly shorter overall survival (OS) and progression-free survival (PFS) times than those with negative TRKC expression (P<0.001). Further, NT-3 expression was significantly associated with lymph node metastasis (P<0.05), distant metastasis (P<0.05), TNM stage (P<0.05), and serum CEA level (P<0.05). The patients with positive NT-3 expression had prolonged OS and PFS compared to those with negative NT-3 expression (P<0.01). The Cox proportional hazards regression model revealed that TRKC expression had a hazard ratio (HR) of 2.679 (P<0.01), while NT-3 expression had a HR of 0.433 (P<0.05). Conclusions We found that the TRKC and NT-3 proteins were closely associated with the occurrence, metastasis, invasion, and tumor marker levels of CRC, and can be used as independent predictors of prognosis in patients with CRC. TRKC mainly indicates a poor prognosis in CRC, while NT-3 indicates a good prognosis.
Collapse
Affiliation(s)
- Shan Chen
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yao Rao
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Weihui Liu
- Department of Oncology, Dazhou Central Hospital, Dazhou, China
| | - Qian Yang
- Department of Oncology, Dazhou Central Hospital, Dazhou, China
| | - Guirong Wu
- Department of Oncology, Dazhou Central Hospital, Dazhou, China
| | - Fudao Wu
- Department of Oncology, Dazhou Central Hospital, Dazhou, China
| | - Youshu Gao
- Department of Oncology, Dazhou Central Hospital, Dazhou, China
| | - Jie Li
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, China
| | - Jiangping Fu
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Oncology, Dazhou Central Hospital, Dazhou, China
| |
Collapse
|
18
|
Althobaiti YS, Alshanbari AH, Alshoaibi DS, Khan EA, Alghamdi HA, Aljuaid SS, Alzilifi AA, Alsulimani NK, Alrizqi OA, Alkhalifa T, Almalki A, Alsanie WF, Gaber A, Alsaab HO. Neurobehavioral and molecular changes in rats exposed to either captagon or counterfeit captagon. Saudi Pharm J 2025; 33:7. [PMID: 40397316 PMCID: PMC12102425 DOI: 10.1007/s44446-025-00007-5] [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: 01/28/2025] [Accepted: 02/26/2025] [Indexed: 05/22/2025] Open
Abstract
The primary ingredient in captagon, a medication that is frequently abused in the Middle East, is fenethylline (FEN), which breaks down into theophylline and amphetamines (AMP). Due to the limited supply of genuine Captagon, fake Captagon (CC) has surfaced, comprising a variety of chemicals such as lidocaine, theophylline, AMP, caffeine, and diphenhydramine. This study compares the neurobehavioral consequences of CC with FEN, emphasizing the higher health concerns associated with CC. A total of 36 male Sprague Dawley rats were split up into five groups: CC (50 or 100 mg/kg), FEN (50 or 100 mg/kg), and control. Following therapy, body temperature and locomotor activity were recorded. Using quantitative real-time PCR (qRT-PCR), the expression of the Brain-Derived Neurotrophic Factor (BDNF) gene was examined in prefrontal cortex (PFC) samples. The findings indicated a higher risk of lethal hyperthermia because CC significantly increased body temperature and locomotor activity in comparison to FEN. Furthermore, a significant reduction in BDNF mRNA levels in the PFC following CC exposure raised the possibility of long-term cognitive and neuroplasticity deficits. According to these results, CC poses a significantly bigger risk because of its unexpected composition and more severe neurobehavioral effects, even though FEN is a recognized social menace. The present study underscores the pressing necessity of public health measures to curb the proliferation and misuse of CC. To lessen these new medications' detrimental effects on people and society as a whole, education about their risks and initiatives to stop their usage are crucial.
Collapse
Affiliation(s)
- Yusuf S Althobaiti
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia.
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia.
- Ministry of Interior, General Administration for Precursors and Laboratories, General Directorate of Narcotics Control, 11564, Riyadh, Saudi Arabia.
| | - Areej H Alshanbari
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Doaa S Alshoaibi
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Eman Abdulghani Khan
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Hanan A Alghamdi
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Saud S Aljuaid
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Ammar A Alzilifi
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Nwaf K Alsulimani
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Osama A Alrizqi
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Turki Alkhalifa
- Ministry of Interior, General Administration for Precursors and Laboratories, General Directorate of Narcotics Control, 11564, Riyadh, Saudi Arabia
| | - Ahmad Almalki
- Ministry of Interior, General Administration for Precursors and Laboratories, General Directorate of Narcotics Control, 11564, Riyadh, Saudi Arabia
| | - Walaa F Alsanie
- Department of Clinical Laboratories Sciences, the Faculty of Applied Medical Sciences, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Ahmed Gaber
- Department of Biology, College of Science, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Hashem O Alsaab
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| |
Collapse
|
19
|
Doroszkiewicz J, Winkel I, Mroczko B. Comparative analysis of neuroinflammatory pathways in Alzheimer's disease, Parkinson's disease, and multiple sclerosis: insights into similarities and distinctions. Front Neurosci 2025; 19:1579511. [PMID: 40364858 PMCID: PMC12069400 DOI: 10.3389/fnins.2025.1579511] [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: 03/31/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Neurodegenerative diseases, contributing to the significant socioeconomic burden due to aging society, are gaining increasing interest. Despite each disease having different etiologies, neuroinflammation is believed to play a crucial role in Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). In addition to the pathogenic function of inflammation in the brain there is growing evidence that immune responses are essential for neuroregeneration. This review compares and contrasts the neuroinflammatory pathways that selected neurodegenerative diseases share and have in common. In AD, tau tangles and beta-amyloid plaques cause microglia and astrocytes to become activated in an inflammatory response. Alpha-synuclein aggregation stimulate neuroinflammation in Parkinson's disease, especially in the substantia nigra. In Multiple Sclerosis an autoimmune attack on myelin is connected to inflammation via invading immune cells. Commonalities include the release of pro-inflammatory mediators like cytokines and activation of signaling pathways such as NF-κB and MAPK. Comprehending these common routes is essential for discovering early diagnostic possibilities for the diseases and possible tailored treatments. Our work underscores the potential for insights into disease mechanisms. Identifying common targets offers promise for advancing our understanding and potential future treatment approaches across these debilitating disorders.
Collapse
Affiliation(s)
- Julia Doroszkiewicz
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, Bialystok, Poland
| | - Izabela Winkel
- Dementia Disorders Centre, Medical University of Wroclaw, Scinawa, Poland
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, Bialystok, Poland
- Department of Biochemical Diagnostics, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
20
|
Taiwo OO, Rehman S, Storey KB. Pancreatic MicroRNAs in Ictidomys tridecemlineatus Associated with Metabolic Diseases: Nature's Insights into Important Biomarkers. Biomolecules 2025; 15:616. [PMID: 40427509 PMCID: PMC12109365 DOI: 10.3390/biom15050616] [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: 02/11/2025] [Revised: 03/31/2025] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
Hibernation involves a profound metabolic rate depression (MRD) that enables certain species to survive prolonged periods of low energy availability. The thirteen-lined ground squirrel uses MRD to arrange cellular and biochemical pathways which suppress nonvital genetic and cellular pathways to conserve internal energy while preserving all essential processes. This study investigates the role of microRNAs (miRNAs) in controlling key signaling pathways and cellular processes in pancreatic tissue during hibernation. Using next-generation sequencing and broad genomic analysis, we analyzed and identified seven differentially expressed miRNAs (miR-29a-3p, miR-22-3p, miR-125-5p, miR-200a-3p, miR-328-3p, miR-21-5p, and miR-148-3p) in the pancreas of hibernating 13-lined ground squirrels (Ictidomys tridecemlineatus). Our findings reveal that these miRNAs regulate pathways involved in glucose homeostasis, including insulin secretion and metabolic regulation, contributing to the unique adaptations of hibernation. These insights advance our understanding of the molecular adaptations underlying hibernation and may have implications for therapeutic strategies targeting metabolic disorders such as diabetes.
Collapse
Affiliation(s)
| | | | - Kenneth B. Storey
- Department of Biology, Carleton University, Ottawa, ON K1S 5B6, Canada; (O.O.T.); (S.R.)
| |
Collapse
|
21
|
Kopalli SR, Behl T, Baldaniya L, Ballal S, Joshi KK, Arya R, Chaturvedi B, Chauhan AS, Verma R, Patel M, Jain SK, Wal A, Gulati M, Koppula S. Neuroadaptation in neurodegenerative diseases: compensatory mechanisms and therapeutic approaches. Prog Neuropsychopharmacol Biol Psychiatry 2025; 139:111375. [PMID: 40280271 DOI: 10.1016/j.pnpbp.2025.111375] [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: 11/26/2024] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
Progressive neuronal loss is a hallmark of neurodegenerative diseases including Alzheimer's, Parkinson's, Huntington's, and Amyotrophic Lateral Sclerosis (ALS), which cause cognitive and motor impairment. Delaying the onset and course of symptoms is largely dependent on neuroadaptation, the brain's ability to restructure in response to damage. The molecular, cellular, and systemic processes that underlie neuroadaptation are examined in this study. These mechanisms include gliosis, neurogenesis, synaptic plasticity, and changes in neurotrophic factors. Axonal sprouting, dendritic remodelling, and compensatory alterations in neurotransmitter systems are important adaptations observed in NDDs; nevertheless, these processes may shift to maladaptive plasticity, which would aid in the advancement of the illness. Amyloid and tau pathology-induced synaptic alterations in Alzheimer's disease emphasize compensatory network reconfiguration. Dopamine depletion causes a major remodelling of the basal ganglia in Parkinson's disease, and non-dopaminergic systems compensate. Both ALS and Huntington's disease rely on motor circuit rearrangement and transcriptional dysregulation to slow down functional deterioration. Neuroadaptation is, however, constrained by oxidative stress, compromised autophagy, and neuroinflammation, particularly in elderly populations. The goal of emerging therapy strategies is to improve neuroadaptation by pharmacologically modifying neurotrophic factors, neuroinflammation, and synaptic plasticity. Neurostimulation, cognitive training, and physical rehabilitation are instances of non-pharmacological therapies that support neuroplasticity. Restoring compensating systems may be possible with the use of stem cell techniques and new gene treatments. The goal of future research is to combine biomarkers and individualized medicines to maximize neuroadaptive responses and decrease the course of illness. In order to reduce neurodegeneration and enhance patient outcomes, this review highlights the dual function of neuroadaptation in NDDs and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Punjab-140306, India
| | - Lalji Baldaniya
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot 360003, Gujarat, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, India; Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Renu Arya
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - Bhumi Chaturvedi
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Ashish Singh Chauhan
- Uttaranchal Institute of Pharmaceutical Sciences, Division of research and innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Rakesh Verma
- Department of Pharmacology, Institute of Medical Science, BHU, Varanasi, India
| | - Minesh Patel
- Department of Pharmacology & Pharmacy Practice, Saraswati Institute of Pharmaceutical Sciences, Dhanap, Gandhinagar, Gujarat, India
| | - Sanmati Kumar Jain
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Koni, Bilaspur, India, 495009
| | - Ankita Wal
- Pranveer Singh Institute of Technology, Pharmacy, NH-19, Bhauti Road, Kanpur, UP, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India; ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| |
Collapse
|
22
|
Ospondpant D, Gao X, Lin S, Ho YM, Dong TTX, Tsim KWK. Pterostilbene Potentiates the NGF-TrkA Signaling Pathway, Enhancing Differentiation in PC12 Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:9595-9605. [PMID: 40204644 DOI: 10.1021/acs.jafc.4c10766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Pterostilbene, a stilbenoid found in vegetables and natural products, has therapeutic potential due to its multiple pharmacological actions. In the brain, the nerve growth factor (NGF) is a pivotal neurotrophic factor, serving for neuronal survival and differentiation. The decline in NGF levels in aged individuals contributes to the development of neurodegenerative processes and cognitive impairment. Here, we aim to explore the effect of pterostilbene on promoting neuron-like differentiation in PC12 cells, a well-established model to study neuronal differentiation, by potentiating the functions of NGF. Molecular docking and ultrafiltration assays were performed to examine the direct binding of pterostilbene with NGF. The mechanisms underlying the stimulation of PC12 cell differentiation, characterized by enhanced neurite outgrowth and increased neurofilament expression, were determined through TrkA/Akt/CREB signaling pathways. The combined treatment of pterostilbene with a low dose of NGF significantly potentiated the NGF-induced neurite extension and neurofilament expression. Pterostilbene enhanced the effect of NGF on promoting neuron-like differentiation, which was related to increased activation of the TrkA signaling pathway. This upstream event was associated with increased phosphorylation of Akt and CREB. The selective inhibitors of TrkA (K252a) and PI3K/Akt (LY294002) were applied to validate the NGF/TrkA/Akt signaling pathways leading to diminished neurite outgrowth and reduced expression of neurofilaments in cells treated with pterostilbene and NGF. Taken together, the results indicate that pterostilbene could potentiate NGF/TrkA activity, enhancing neuron-like differentiation in PC12 cells under a low concentration of NGF. These findings suggest that the application of pterostilbene could be a promising alternative therapeutic strategy to improve NGF efficacy.
Collapse
Affiliation(s)
- Dusadee Ospondpant
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong 999077, China
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, Shenzhen Research Institute, The Hong Kong University of Science and Technology, Shenzhen 518000, China
| | - Xiong Gao
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong 999077, China
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, Shenzhen Research Institute, The Hong Kong University of Science and Technology, Shenzhen 518000, China
| | - Shengying Lin
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong 999077, China
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, Shenzhen Research Institute, The Hong Kong University of Science and Technology, Shenzhen 518000, China
| | - Yuen Man Ho
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong 999077, China
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, Shenzhen Research Institute, The Hong Kong University of Science and Technology, Shenzhen 518000, China
| | - Tina Ting Xia Dong
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong 999077, China
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, Shenzhen Research Institute, The Hong Kong University of Science and Technology, Shenzhen 518000, China
| | - Karl Wah Keung Tsim
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong 999077, China
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, Shenzhen Research Institute, The Hong Kong University of Science and Technology, Shenzhen 518000, China
| |
Collapse
|
23
|
Grosu-Bularda A, Vancea CV, Hodea FV, Cretu A, Bordeanu-Diaconescu EM, Dumitru CS, Ratoiu VA, Teodoreanu RN, Lascar I, Hariga CS. Optimizing Peripheral Nerve Regeneration: Surgical Techniques, Biomolecular and Regenerative Strategies-A Narrative Review. Int J Mol Sci 2025; 26:3895. [PMID: 40332790 PMCID: PMC12027958 DOI: 10.3390/ijms26083895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/09/2025] [Accepted: 04/18/2025] [Indexed: 05/08/2025] Open
Abstract
Peripheral nerve injury disrupts the function of the peripheral nervous system, leading to sensory, motor, and autonomic deficits. While peripheral nerves possess an intrinsic regenerative capacity, complete sensory and motor recovery remains challenging due to the unpredictable nature of the healing process, which is influenced by the extent of the injury, age, and timely intervention. Recent advances in microsurgical techniques, imaging technologies, and a deeper understanding of nerve microanatomy have enhanced functional outcomes in nerve repair. Nerve injury initiates complex pathophysiological responses, including Wallerian degeneration, macrophage activation, Schwann cell dedifferentiation, and axonal sprouting. Complete nerve disruptions require surgical intervention to restore nerve continuity and function. Direct nerve repair is the gold standard for clean transections with minimal nerve gaps. However, in cases with larger nerve gaps or when direct repair is not feasible, alternatives such as autologous nerve grafting, vascularized nerve grafts, nerve conduits, allografts, and nerve transfers may be employed. Autologous nerve grafts provide excellent biocompatibility but are limited by donor site morbidity and availability. Vascularized grafts are used for large nerve gaps and poorly vascularized recipient beds, while nerve conduits serve as a promising solution for smaller gaps. Nerve transfers are utilized when neither direct repair nor grafting is possible, often involving re-routing intact regional nerves to restore function. Nerve conduits play a pivotal role in nerve regeneration by bridging nerve gaps, with significant advancements made in material composition and design. Emerging trends in nerve regeneration include the use of 3D bioprinting for personalized conduits, gene therapy for targeted growth factor delivery, and nanotechnology for nanofiber-based conduits and stem cell therapy. Advancements in molecular sciences have provided critical insights into the cellular and biochemical mechanisms underlying nerve repair, leading to targeted therapies that enhance axonal regeneration, remyelination, and functional recovery in peripheral nerve injuries. This review explores the current strategies for the therapeutic management of peripheral nerve injuries, highlighting their indications, benefits, and limitations, while emphasizing the need for tailored approaches based on injury severity and patient factors.
Collapse
Affiliation(s)
- Andreea Grosu-Bularda
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Cristian-Vladimir Vancea
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Florin-Vlad Hodea
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Andrei Cretu
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Eliza-Maria Bordeanu-Diaconescu
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Catalina-Stefania Dumitru
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Vladut-Alin Ratoiu
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Razvan-Nicolae Teodoreanu
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Ioan Lascar
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Cristian-Sorin Hariga
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| |
Collapse
|
24
|
Radhakrishna U, Radhakrishnan R, Uppala LV, Trivedi TS, Prajapati J, Rawal RM, Muvvala SB, Bahado-Singh RO, Sadhasivam S. Prenatal opioid exposure alters pain perception and increases long-term health risks in infants with neonatal opioid withdrawal syndrome. FRONTIERS IN PAIN RESEARCH 2025; 6:1497801. [PMID: 40313396 PMCID: PMC12043715 DOI: 10.3389/fpain.2025.1497801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 03/24/2025] [Indexed: 05/03/2025] Open
Abstract
Background Opioids are often prescribed for pain relief, yet they pose risks such as addiction, dependence, and overdose. Pregnant women have unique vulnerabilities to opioids and infants born to opioid-exposed mothers could develop neonatal opioid withdrawal syndrome (NOWS). The study of opioid-induced epigenetic changes in chronic pain is in its early stages. This study aimed to identify epigenetic changes in genes associated with chronic pain resulting from maternal opioid exposure during pregnancy. Methods We analyzed DNA methylation of chronic pain-related genes in 96 placental tissues using Illumina Infinium Methylation EPIC BeadChips. These samples comprised 32 from mothers with infants prenatally exposed to opioids who needed pharmacologic NOWS management (+Opioids/+NOWS), 32 from mothers with prenatally opioid-exposed infants not needing NOWS pharmacologic treatment (+Opioids/-NOWS), and 32 from unexposed control subjects (-Opioids/-NOWS). Results The study identified significant methylation changes at 111 CpG sites in pain-related genes among opioid-exposed infants, with 54 CpGs hypomethylated and 57 hypermethylated. These genes play a crucial role in various biological processes, including telomere length regulation (NOS3, ESR1, ESR2, MAPK3); inflammation (TNF, MAPK3, IL1B, IL23R); glucose metabolism (EIF2AK3, CACNA1H, NOTCH3, GJA1); ion channel function (CACNA1C, CACNA1H, CLIC4, KCNQ5); autophagy (CTSS, ULK1, ULK4, ATG5); oxidative stress (NGF, NRG1, OPRM1, ATP1A2); aging (GRIA1, NGFR, PRLR, EIF4E); cytokine activity (TRPV4, RUNX1, CXCL8, IL18R1); and the risk of suicide (ADORA2A, ANKK1, GABRG2, IGSF9B). These epigenetic changes may influence 48 signaling pathways-including cAMP, MAPK, GnRH secretion, estrogen signaling, morphine addiction, circadian rhythms, and insulin secretion-profoundly affecting pain and inflammation-related processes. Conclusion The identified methylation alterations may shed light on pain, neurodevelopmental changes, and other biological mechanisms in opioid-exposed infants and mothers with OUD, offering insights into NOWS and maternal-infant health. These findings may also pave the way for targeted interventions and improved pain management, highlighting the potential for integrated care strategies to address the interconnected health of mothers and infants.
Collapse
Affiliation(s)
- Uppala Radhakrishna
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Obstetrics and Gynecology, Corewell Health William Beaumont University Hospital, Royal Oak, MI, United States
| | - Rupa Radhakrishnan
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Lavanya V. Uppala
- Department of Pharmacology & Neuroscience, School of Medicine, Creighton University, Omaha, NE, United States
| | - Tithi S. Trivedi
- Department of Botany, Bioinformatics and Climate Change Impacts Management, School of Sciences, Gujarat University, Ahmedabad, Gujarat, India
| | - Jignesh Prajapati
- Department of Biochemistry & Forensic Sciences, Gujarat University, Ahmedabad, India
| | - Rakesh M. Rawal
- Department of Medical Biotechnology, Gujarat Biotechnology University, Gandhinagar, Gujarat, India
| | - Srinivas B. Muvvala
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, United States
| | - Ray O. Bahado-Singh
- Department of Obstetrics and Gynecology, Corewell Health William Beaumont University Hospital, Royal Oak, MI, United States
| | - Senthilkumar Sadhasivam
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
25
|
Kılıç KD, Garipoğlu G, Çakar B, Uyanıkgil Y, Erbaş O. Antioxidant-Effective Quercetin Through Modulation of Brain Interleukin-13 Mitigates Autistic-Like Behaviors in the Propionic Acid-Induced Autism Model in Rats. J Neuroimmune Pharmacol 2025; 20:36. [PMID: 40220083 PMCID: PMC11993503 DOI: 10.1007/s11481-025-10190-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/20/2025] [Indexed: 04/14/2025]
Abstract
Overproduction of reactive oxygen species occurs when inflammation induces oxidative stress in macrophages and microglia, leading to a self-sustaining cycle of cellular damage and neuroinflammation. Oxidative stress and neuroinflammation are well-established contributors to the pathophysiology of autism spectrum disorders, which are associated with impaired neuronal function, neuronal loss, and behavioral deficits. Damaged cells, through microglial activation, release additional inflammatory mediators under conditions of oxidative stress, exacerbating neuronal damage. Quercetin, a powerful dietary antioxidant, has been shown to scavenge free radicals, reduce oxidative stress, and inhibit inflammatory pathways. Given these properties, we hypothesize that quercetin may improve learning and social skills in individuals with autism spectrum disorders by alleviating oxidative stress and reducing brain levels of inflammatory cytokines. In this study, an autism model was established in 30 rats by intraperitoneal injection of 250 mg/kg/day propionic acid (PPA) for five days. The study groups were as follows: Group 1: Normal ontrol (n = 10); Group 2: PPA + saline (PPAS, n = 10); Group 3: PPA + Quercetin (PPAQ, n = 10). All treatments were administered for 15 days. At the end of the treatment, histological and biochemical analyses of brain tissue and behavioral tests related to autistic-like behaviors were performed. Malondialdehyde, tumor necrosis factor-alpha, and interleukin-13 levels in brain homogenates were significantly higher in the PPAS group compared to the control group, indicating elevated oxidative stress and inflammation following PPA exposure. The PPAQ group significantly reduced oxidative stress parameters and inflammatory biomarkers, demonstrating its antioxidant and anti-inflammatory effects. This biochemical improvement was accompanied by preserving Purkinje cells and neuronal populations, significantly reduced in the PPAS group. Moreover, quercetin-treated rats exhibited improved social behavior and learning, which were severely impaired in the PPAS group. These findings, when interpreted together, suggest that quercetin exerts its neuroprotective effects by targeting oxidative stress and neuroinflammation, thereby preventing neuronal cell loss and alleviating behavioral deficits associated with autism spectrum disorders.
Collapse
Affiliation(s)
- Kubilay Doğan Kılıç
- Faculty of Medicine, Department of Histology and Embryology, Ege University, İzmir, Türkiye.
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Zentrum München, Munich, Germany.
- Museum Für Naturkunde, Leibniz Institute for Evolution and Biodiversity Science, Berlin, Germany.
| | - Gökçen Garipoğlu
- Faculty of Health Sciences, Department of Nutrition and Dietetic, Bahçeşehir University, Istanbul, Türkiye
| | - Burak Çakar
- Faculty of Medicine, Department of Histology and Embryology, İstinye University, İstanbul, Türkiye
| | - Yiğit Uyanıkgil
- Faculty of Medicine, Department of Histology and Embryology, Ege University, İzmir, Türkiye
- Cord Blood Cell - Tissue Research and Application Center, Ege University, İzmir, Türkiye
| | - Oytun Erbaş
- Faculty of Medicine, Biruni Research Center (BAMER), Biruni University, Istanbul, Türkiye
| |
Collapse
|
26
|
Bhavsar K, Tripathi M, Banerjee J, Srivastava A, Nidhi, Pandey S, Vohora D. Brain-derived neurotrophic factor but not beta-secretase 1, vascular endothelial growth factor, glial fibrillary acidic protein and interleukin-1β correlate with cognitive impairment in adult persons with epilepsy: a cross-sectional single-center study from India. Front Neurol 2025; 16:1540915. [PMID: 40291844 PMCID: PMC12023754 DOI: 10.3389/fneur.2025.1540915] [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: 12/06/2024] [Accepted: 02/14/2025] [Indexed: 04/30/2025] Open
Abstract
Objectives This study aims to evaluate cognitive impairment utilizing the Montreal Cognitive Assessment (MoCA) scale, while also exploring the correlation between cognitive impairment and various serum biomarkers, including Brain-derived neurotrophic factor (BDNF), Beta Secretase-1 (BACE1), Vascular Endothelial Growth Factors (VEGF), Glial fibrillary acidic protein (GFAP), and Interleukin-1 (IL-1β) in adults living with epilepsy. Methods In this study, 74 participants aged between 18 and 50 years, who were visiting neurology outpatient consultations, were included. The cognitive assessment was executed using the MoCA test. Serum levels of BDNF, BACE1, VEGF, GFAP, and IL-1β were evaluated through ELISA in patients with and without cognitive impairments. To determine the association between MoCA scores and the biomarkers, both Spearman and Pearson correlation analyses, as well as linear regression, were conducted. Results Among the 74 PWE, 61 exhibited cognitive impairment as determined by the MoCA assessment. Noteworthy alterations were detected across various MoCA subscales, encompassing visuospatial and executive functions, attention, language, abstraction, and delayed recall, with statistical significance established (p < 0.05). Furthermore, it was revealed that those in the cognitively impaired group presented with reduced serum BDNF levels (p < 0.05). It is important to highlight that no substantial differences were identified in the serum concentrations of BACE-1, VEGF, GFAP, and IL-1β. A moderate and statistically significant correlation was established between BDNF and the Total MoCA score (p < 0.05), in addition to BDNF's relationship with Visuospatial & Executive function (p < 0.05). In the context of regression analysis, BDNF demonstrated a significant association to the Total MoCA score (p < 0.05), a connection that persisted as significant even when adjusted for confounding factors. Conclusion We conclude that adult PWE in India demonstrate a significant cognitive impairment. Further, our findings indicate that BDNF may serve as a potential biomarker for evaluating cognitive impairment in adult PWE. Further longitudinal, prospective and multi-center studies are required to confirm the same.
Collapse
Affiliation(s)
- Kamini Bhavsar
- Department of Pharmacology, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, India
| | - Manjari Tripathi
- Department of Neurology, All India Institute of Medical Science (AIIMS), New Delhi, India
| | - Jyotirmoy Banerjee
- Department of Biophysics, All India Institute of Medical Science (AIIMS), New Delhi, India
| | - Arpna Srivastava
- Department of Neurology, All India Institute of Medical Science (AIIMS), New Delhi, India
| | - Nidhi
- School of Chemical and Life Sciences (SCLS), Jamia Hamdard, New Delhi, India
| | - Shivam Pandey
- Department of Biostatistics, All India Institute of Medical Science (AIIMS), New Delhi, India
| | - Divya Vohora
- Department of Pharmacology, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, India
| |
Collapse
|
27
|
Madhubala D, Mahato R, Saikia K, Patra A, Fernandes PA, Kumar A, Khan MR, Mukherjee AK. Snake Venom-Inspired Novel Peptides Protect Caenorhabditis elegans against Paraquat-Induced Parkinson's Pathology. ACS Chem Neurosci 2025; 16:1275-1296. [PMID: 40096006 DOI: 10.1021/acschemneuro.4c00576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025] Open
Abstract
The in vivo protective mechanisms of two low-molecular-mass (∼1.4 kDa) novel custom peptides (CPs) against paraquat-induced neurodegenerative dysfunction in the Caenorhabditis elegans model were deciphered. CPs prevented the paraquat from binding to the nerve ring adjacent to the pharynx in C. elegans (wild-type) by stable and high-affinity binding to the tyrosine-protein kinase receptor CAM-1, resulting in significant inhibition of paraquat-induced toxicity by reducing the production of reactive oxygen species, mitochondrial membrane depolarization, and chemosensory dysfunction. The CPs inhibited paraquat-induced dopaminergic neuron degeneration and alpha-synuclein protein expression, the hallmarks of Parkinson's disease, in transgenic BZ555 and NL5901 strains of C. elegans. Transcriptomic, functional proteomics, and quantitative reverse transcription-polymerase chain reaction analyses show that CPs prevented the increased expression of the genes involved in the skn-1 downstream pathway, thereby restoring paraquat-mediated oxidative stress, apoptosis, and neuronal damage in C. elegans. The ability of CPs to repair paraquat-induced damage was demonstrated by a network of gene expression profiles, illustrating the molecular relationships between the regulatory proteins.
Collapse
Affiliation(s)
- Dev Madhubala
- Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur, Assam 784028, India
- Institute of Advanced Studies in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati, Assam 781035, India
| | - Rosy Mahato
- Institute of Advanced Studies in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati, Assam 781035, India
- Faculty of Science, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Kangkon Saikia
- Institute of Advanced Studies in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati, Assam 781035, India
| | - Aparup Patra
- Institute of Advanced Studies in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati, Assam 781035, India
| | - Pedro Alexandrino Fernandes
- LAQV@REQUIMTE, Departamento de Química e Bioquímica, Faculdade De Ciências, Universidade do Porto, Rua Do Campo Alegre S/N, Porto 4169-007, Portugal
| | - Arun Kumar
- Institute of Advanced Studies in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati, Assam 781035, India
| | - Mojibur R Khan
- Institute of Advanced Studies in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati, Assam 781035, India
| | - Ashis K Mukherjee
- Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur, Assam 784028, India
- Institute of Advanced Studies in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati, Assam 781035, India
| |
Collapse
|
28
|
Wu H, Zhou JJ, Chen XY, Zhu DD, Bao F, Zheng W, Ren L, Pan WG, Liu CM. BDNF Gene Polymorphism and Antidepressant Response in Han Chinese Patients with First-Episode Late-Life Depression. ALPHA PSYCHIATRY 2025; 26:39955. [PMID: 40352065 PMCID: PMC12059767 DOI: 10.31083/ap39955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/13/2024] [Accepted: 10/31/2024] [Indexed: 05/14/2025]
Abstract
Objective This study investigated the association between brain-derived neurotrophic factor (BDNF) gene polymorphisms and antidepressant response in patients with first-episode late-life depression (LLD). Methods A total of 72 patients with first-episode LLD were recruited and 57 completed an 8-week course of antidepressant treatment. Participants were assessed at baseline and post-treatment using the 17-item Hamilton Depression Rating Scale (HAMD-17) and the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS). Serum BDNF levels were measured via Enzyme-Linked Immunosorbent Assay (ELISA) and BDNF gene polymorphisms were genotyped using the Agena® MassARRAY system. Results After 8 weeks, 17 of the 57 patients with LLD showed effective treatment response (effective group), while 40 were classified as ineffective. Significant post-treatment improvements were observed across the cohort in HAMD-17 and RBANS scores, and serum BDNF levels compared with baseline (p < 0.05). However, the effective and ineffective groups did not have significantly different RBANS scores or serum BDNF levels (p > 0.05). Binary logistic regression identified male sex (OR = 10.094, p = 0.007) and BDNF gene polymorphism (OR = 6.559, p = 0.003) as predictors of treatment efficacy. Conclusion Antidepressant treatment for 8 weeks altered serum BDNF levels in patients with LLD, with male patients carrying the Val/Val genotype potentially responded better to conventional antidepressants. The small sample size may limit the generalizability of these findings. Clinical Trial Registration The study was registered at https://www.chictr.org.cn (registration number: ChiCTR1900024445).
Collapse
Affiliation(s)
- Han Wu
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, 100088 Beijing, China
| | - Jiao-jiao Zhou
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, 100088 Beijing, China
| | - Xue-yan Chen
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, 100088 Beijing, China
| | - Dan-di Zhu
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, 100088 Beijing, China
| | - Feng Bao
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, 100088 Beijing, China
| | - Wei Zheng
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, 510370 Guangzhou, Guangdong, China
| | - Li Ren
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, 100088 Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, 100088 Beijing, China
| | - Wei-gang Pan
- Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, 100096 Beijing, China
| | - Chao-meng Liu
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, 100088 Beijing, China
| |
Collapse
|
29
|
Jafari A, Khoshdooz S, Bafrani MA, Bakhshimoghaddam F, Abbasi H, Doaei S. Uncovering the Causal Link Between Obesity-Associated Genes and Multiple Sclerosis: A Systematic Literature Review. Brain Behav 2025; 15:e70439. [PMID: 40195065 PMCID: PMC11975544 DOI: 10.1002/brb3.70439] [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: 10/11/2024] [Revised: 02/25/2025] [Accepted: 03/02/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a multifaceted neurodegenerative disorder influenced by genetics and lifestyle. This systematic literature review investigates the role of six obesity-associated genes, including fat mass and obesity-associated (FTO), FAS apoptosis inhibitory molecule 2 (FAIM2), Niemann-Pick disease type C1-like 1 (NPC1), glucosamine-6-phosphate deaminase 2 (GNPDA2), melanocortin-4 receptor (MC4R), and brain-derived neurotrophic factor (BDNF) in the context of MS. METHODS A literature search was executed using Embase, Scopus, Cochrane, Web of Science, and PubMed databases from inception to July 2024. The related keywords employed during the search process are "fas apoptotic inhibitory molecule 2," "Niemann-Pick disease type C1," "fat mass and obesity-associated," "melanocortin-4 receptor," "brain-derived neurotrophic factor," "glucosamine-6-phosphate deaminase 2," and "multiple sclerosis." RESULTS Out of 2108 papers, 27 were entered into the present systematic literature review. The FTO gene may affect MS susceptibility through metabolic and inflammatory pathways. FAIM2 and NPC1 genes may contribute to MS pathogenesis, though their precise roles are still being elucidated. The GNPDA2 gene may have some connections with MS but requires further clarification. MC4R has demonstrated significant neuroprotective and anti-inflammatory effects, suggesting its potential impact on MS progression. BDNF plays a complex role in neuronal survival and repair and may influence the risk of MS. CONCLUSION Our findings demonstrated that obesity-related genes may have a significant impact on MS risk and disease course, revealing novel insights into the genetic underpinnings of MS.
Collapse
Affiliation(s)
- Ali Jafari
- Student Research Committee, Department of Community Nutrition, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research InstituteShahid Beheshti University of Medical SciencesTehranIran
- Systematic Review and Meta‐analysis Expert Group (SRMEG)Universal Scientific Education and Research Network (USERN)TehranIran
| | - Sara Khoshdooz
- Faculty of MedicineGuilan University of Medical ScienceRashtIran
| | | | - Farnush Bakhshimoghaddam
- Department of Nutrition, School of Allied Medical SciencesAhvaz Jundishapur University of Medical SciencesAhvazIran
| | - Hamid Abbasi
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
- Neurosciences Research CenterTabriz University of Medical SciencesTabrizIran
| | - Saeid Doaei
- Department of Community Nutrition, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research InstituteShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
30
|
Elahinejad V, Khorasanian AS, Tehrani‐Doost M, Khosravi‐Darani K, Mirsepasi Z, Effatpanah M, Askari‐Rabori R, Tajadod S, Jazayeri S. Effects of Probiotics as Adjunctive Therapy to Fluoxetine on Depression Severity and Serum Brain-Derived Neurotrophic Factor, Cortisol, and Adrenocorticotropic Hormone in Patients With Major Depressive Disorder: A Randomized, Double-Blind, Placebo-Controlled Trial. Food Sci Nutr 2025; 13:e4698. [PMID: 40177327 PMCID: PMC11961381 DOI: 10.1002/fsn3.4698] [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: 08/17/2024] [Revised: 11/22/2024] [Accepted: 12/03/2024] [Indexed: 04/05/2025] Open
Abstract
Probiotics may improve mood, but their role as adjunctive therapy for major depressive disorder (MDD) is not well understood. This study examines the effects of probiotics on depression severity, brain-derived neurotrophic factor (BDNF), adrenocorticotropic hormone (ACTH), and cortisol levels in MDD patients. Fifty medication-free MDD patients were randomized to receive probiotics with fluoxetine (n = 25) or placebo with fluoxetine (n = 25) for 8 weeks. Depression severity was assessed using the Hamilton Depression Rating Scale (HDRS-24), and fasting blood samples were collected at baseline and study conclusion. Forty-four patients completed the trial. The probiotic group showed a significant reduction in depression severity compared with the placebo group (p = 0.001). No significant differences were observed in serum cortisol (p = 0.46) and ACTH levels (p = 0.44). Plasma BDNF levels increased slightly in the probiotic group but were not statistically significant. Probiotic supplementation with fluoxetine significantly reduces depression severity in MDD patients.
Collapse
Affiliation(s)
- Vajihe Elahinejad
- Department of Nutrition, School of Public HealthIran University of Medical SciencesTehranIran
| | - Atie Sadat Khorasanian
- Department of Nutrition, School of Public HealthIran University of Medical SciencesTehranIran
- Endocrinology and Metabolism Research CenterEndocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical SciencesTehranIran
| | - Mehdi Tehrani‐Doost
- Department of PsychiatryRoozbeh Hospital, Tehran University of Medical SciencesTehranIran
| | - Kianoush Khosravi‐Darani
- Research Department of Food Technology ResearchNational Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical SciencesTehranIran
| | - Zahra Mirsepasi
- Department of PsychiatryRoozbeh Hospital, Tehran University of Medical SciencesTehranIran
| | - Mohammad Effatpanah
- School of MedicineZiaeian Hospital, International Campus, Tehran University of Medical SciencesTehranIran
| | | | - Shirin Tajadod
- Department of Nutrition, School of Public HealthIran University of Medical SciencesTehranIran
| | - Shima Jazayeri
- Department of Nutrition, School of Public HealthIran University of Medical SciencesTehranIran
- Research Center for Nutritional SciencesIran University of Medical SciencesTehranIran
| |
Collapse
|
31
|
Tao Y, Yi X, Zhou X, Qu J, Diogene T, Wang A, Zhang Y. Link between gut damage and neurotoxicity with gender differences in zebrafish: Dibutyl phthalate-driven microbiota dysbiosis as a possible major cause. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 972:179102. [PMID: 40090240 DOI: 10.1016/j.scitotenv.2025.179102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/18/2025]
Abstract
Among plasticizers, dibutyl phthalate (DBP) is widely used in in industry, posing significant health risks to aquatic organisms. In this study, adult male and female zebrafish were exposed to 0 and 30 μg/L DBP for 15 days. Behavioral monitoring, immunofluorescence, protein immunoblotting, and high-throughput sequencing were used to investigate the critical role of the gut microbiome in DBP-induced dysfunction of the zebrafish gut-brain axis. The results showed pronounced, sex-specific toxic effects of acute DBP exposure in adult zebrafish, with males experiencing more severe neurological damage, while females exhibited greater intestinal damage. DBP exposure caused marked anxiety behaviors in males and significant weight loss in females. Males showed reduced neuronal expression, while females exhibited increased intestinal permeability and lower levels of the tight junction protein (ZO-1). The Firmicutes/Bacteroidota (F/B) ratio decreased, indicating severe gut microbiota dysbiosis. Changes in the gut and fecal microbiota composition, along with PICRUSt2 functional predictions, suggest that female zebrafish experienced more severe metabolic disturbances than males. Analysis of key gene expression in the brain-derived neurotrophic factor (bdnf) pathway revealed that changes in the abundance of tryptophan-metabolizing bacteria in the gut may explain the sex-specific effects of DBP on neurotransmitter serotonin levels in the brain, which influence the gut-brain axis in zebrafish. This study contributes to the understanding of toxic effects of DBP on aquatic organisms and provides strong evidence for assessing its environmental risks.
Collapse
Affiliation(s)
- Yue Tao
- School of Resources and Environment, Northeast Agricultural University, Harbin 150030, PR China
| | - Xiaodong Yi
- School of Resources and Environment, Northeast Agricultural University, Harbin 150030, PR China
| | - Xinyi Zhou
- School of Resources and Environment, Northeast Agricultural University, Harbin 150030, PR China
| | - Jianhua Qu
- School of Resources and Environment, Northeast Agricultural University, Harbin 150030, PR China
| | - Tuyiringire Diogene
- School of Resources and Environment, Northeast Agricultural University, Harbin 150030, PR China
| | - Aoxue Wang
- School of Horticulture and Landscape Architecture, Northeast Agricultural University, Harbin 150030, PR China
| | - Ying Zhang
- School of Resources and Environment, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
32
|
Trigo CM, Rodrigues JS, Camões SP, Solá S, Miranda JP. Mesenchymal stem cell secretome for regenerative medicine: Where do we stand? J Adv Res 2025; 70:103-124. [PMID: 38729561 PMCID: PMC11976416 DOI: 10.1016/j.jare.2024.05.004] [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: 08/15/2023] [Revised: 02/27/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Mesenchymal stem cell (MSC)-based therapies have yielded beneficial effects in a broad range of preclinical models and clinical trials for human diseases. In the context of MSC transplantation, it is widely recognized that the main mechanism for the regenerative potential of MSCs is not their differentiation, with in vivo data revealing transient and low engraftment rates. Instead, MSCs therapeutic effects are mainly attributed to its secretome, i.e., paracrine factors secreted by these cells, further offering a more attractive and innovative approach due to the effectiveness and safety of a cell-free product. AIM OF REVIEW In this review, we will discuss the potential benefits of MSC-derived secretome in regenerative medicine with particular focus on respiratory, hepatic, and neurological diseases. Both free and vesicular factors of MSC secretome will be detailed. We will also address novel potential strategies capable of improving their healing potential, namely by delivering important regenerative molecules according to specific diseases and tissue needs, as well as non-clinical and clinical studies that allow us to dissect their mechanisms of action. KEY SCIENTIFIC CONCEPTS OF REVIEW MSC-derived secretome includes both soluble and non-soluble factors, organized in extracellular vesicles (EVs). Importantly, besides depending on the cell origin, the characteristics and therapeutic potential of MSC secretome is deeply influenced by external stimuli, highlighting the possibility of optimizing their characteristics through preconditioning approaches. Nevertheless, the clarity around their mechanisms of action remains ambiguous, whereas the need for standardized procedures for the successful translation of those products to the clinics urges.
Collapse
Affiliation(s)
- Catarina M Trigo
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana S Rodrigues
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Sérgio P Camões
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Susana Solá
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana P Miranda
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
33
|
Su H, Ye Q, Wang D, Liu A, Wang Y, Zhang Y, Weng L. A self-assembling peptide-based hydrogel containing NF- κB inhibitors and NGF for peripheral nerve injury repair. Biofabrication 2025; 17:025031. [PMID: 40145273 DOI: 10.1088/1758-5090/adc340] [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: 10/26/2024] [Accepted: 03/20/2025] [Indexed: 03/28/2025]
Abstract
An inflammatory response may be initiated after peripheral nerve injury (PNI), potentially hindering the repair and regeneration of damaged nerves. Administering anti-inflammatory agents to modulate macrophage phenotypes may reduce post-injury inflammation and show potential for treating PNI. Regrettably, the limited half-lives of these compounds within the human body constrain their efficacy as anti-inflammatory agents. In this study, we co-assembled picroside II (PII) and nerve growth factor (NGF) with the hydrogelator compound Nap-Phe-Phe-Tyr-OH (NapFFY) to form a supramolecular hydrogel, PII/NGF/NapFFY@Gel, which could be accurately delivered to the nerve injury site viain situinjection to improve its bioavailability. Our results demonstrated that the PII/NGF/NapFFY@Gel exhibits favorable drug slow-release performance in bothin vivoandin vitroexperiments. Furthermore, cell and animal studies revealed that the PII/NGF/NapFFY@Gel effectively enhanced nerve recovery and regeneration by modulating the inflammatory microenvironment. This mechanism involves inhibiting the NF-κB inflammatory signaling pathway, suppressing macrophage polarization to the M1 phenotype, and upregulating the expression of proteins associated with nerve regeneration. Taken together, the results of this study suggest that improving the inflammatory microenvironment and promoting nerve repair through thein situinjection of PII/NGF/NapFFY@Gel with sustained drug release may be a novel treatment for PNI.
Collapse
Affiliation(s)
- HaiTao Su
- Department of Pain, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, People's Republic of China
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, People's Republic of China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, 678 Furong Road, Hefei 230601, People's Republic of China
| | - QiuPing Ye
- Department of Pain, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, People's Republic of China
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, People's Republic of China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, 678 Furong Road, Hefei 230601, People's Republic of China
| | - Di Wang
- Department of Pain, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, People's Republic of China
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, People's Republic of China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, 678 Furong Road, Hefei 230601, People's Republic of China
| | - AnQi Liu
- Department of Pain, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, People's Republic of China
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, People's Republic of China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, 678 Furong Road, Hefei 230601, People's Republic of China
| | - YaGuang Wang
- Department of Pain, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, People's Republic of China
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, People's Republic of China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, 678 Furong Road, Hefei 230601, People's Republic of China
| | - Ye Zhang
- Department of Pain, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, People's Republic of China
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, People's Republic of China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, 678 Furong Road, Hefei 230601, People's Republic of China
| | - LiJun Weng
- Department of Pain, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, People's Republic of China
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, People's Republic of China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, 678 Furong Road, Hefei 230601, People's Republic of China
| |
Collapse
|
34
|
Petrovici A, Savuța G, Lucini C, Robea MA, Solcan C. Combined Neurotoxic Effects of Commercial Formulations of Pyrethroid (Deltamethrin) and Neonicotinoid (Imidacloprid) Pesticides on Adult Zebrafish ( Danio rerio): Behavioral, Molecular, and Histopathological Analysis. Life (Basel) 2025; 15:538. [PMID: 40283093 PMCID: PMC12028659 DOI: 10.3390/life15040538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 04/29/2025] Open
Abstract
The use of different commercial products that involve one or multiple active substances with specific targeted-pests control has become a widespread practice. Because of this, a severe range of significant consequences has been often reported. Among the most used pesticides worldwide are deltamethrin (DM) and imidacloprid (IMI). With a significative effect on the insect's nervous system, DM acts on the voltage-gated sodium channels in nerve cell membranes, while IMI mimics the acetylcholine neurotransmitter by binding irreversibly to the nicotinic acetylcholine receptors. This study investigates the neurotoxic effects of sub-chronic exposure to commercial formulations of deltamethrin (DM) and imidacloprid (IMI) in adult zebrafish, both individually and in combination. The formulations used in this study contain additional ingredients commonly found in commercial pesticide products, which may contribute to overall toxicity. Fish were exposed to environmentally relevant concentrations of these pesticides for 21 days, individually or in combination. Behavioral, molecular, and histopathological analyses were conducted to assess the impact of these pesticides. Zebrafish exhibited dose-dependent behavioral alterations, particularly in the combined exposure groups, including increased erratic swimming and anxiety-like behavior. Gene expression analysis revealed significant changes in neurotrophic factors (BDNF, NGF, ntf-3, ntf-4/5, ntf-6/7) and their receptors (ntrk1, ntrk2a, ntrk2b, ntrk3a, ntrk3b, ngfra, ngfrb), indicating potential neurotoxic effects. Histopathological examination confirmed neuronal degeneration, gliosis, and vacuolization, with more severe impairments observed in pesticide mixture treatments. These findings highlight the neurotoxic potential of pesticide formulations in aquatic environments and emphasize the need for stricter regulations on pesticide mixtures and further research on pesticide interactions. Our findings emphasize that the combination of pesticides could trigger a synergistic effect by maximizing the toxicity of each compound. Thus, it is a well-known practice for pyrethroids and neonicotinoids to be used together in agriculture. Even so, its prevalence in agriculture and the need to investigate its actual impact on human health, biodiversity, and ecosystem mitigates the development of new strategies for assessing the risk and, at the same time, enhancing the effectiveness.
Collapse
Affiliation(s)
- Adriana Petrovici
- Faculty of Veterinary Medicine, “Ion Ionescu de la Brad” Iasi University of Life Sciences (IULS), 8 Mihail Sadoveanu Alley, 700489 Iasi, Romania; (A.P.); (G.S.)
| | - Gheorghe Savuța
- Faculty of Veterinary Medicine, “Ion Ionescu de la Brad” Iasi University of Life Sciences (IULS), 8 Mihail Sadoveanu Alley, 700489 Iasi, Romania; (A.P.); (G.S.)
| | - Carla Lucini
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Napoli, Italy
| | - Madalina-Andreea Robea
- Department of Biology, Doctoral School of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iasi, Bd. Carol I, 20 A, 700505 Iasi, Romania;
| | - Carmen Solcan
- Faculty of Veterinary Medicine, “Ion Ionescu de la Brad” Iasi University of Life Sciences (IULS), 8 Mihail Sadoveanu Alley, 700489 Iasi, Romania; (A.P.); (G.S.)
| |
Collapse
|
35
|
Zheng X, Liu X, Guo Y, Lv Y, Lin C, Wang D, Wang S, Liu Y, Hu X. Physical exercise and epigenetic modifications in skeletal muscle, brain, and heart. Epigenetics Chromatin 2025; 18:12. [PMID: 40114219 PMCID: PMC11927307 DOI: 10.1186/s13072-025-00576-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 02/21/2025] [Indexed: 03/22/2025] Open
Abstract
The origins of many diseases can be traced to the dynamic interplay of genetic predispositions and environmental exposures post-birth. Epigenetic modifications have recently gained prominence as a significant mediator between genetic information and environmental factors, influencing the occurrence and progression of disease. There is a burgeoning body of evidence supports that physical exercise, acting as an external environmental stimulus, exerts a discernible impact on major epigenetic modifications, including histone modifications, DNA methylation, RNA methylation, and non-coding RNA. This effect assumes a pivotal role in the pathogenesis of various human diseases. Exploring the epigenetic molecular mechanisms through which physical exercise enhances human health holds the promise of deepening our understanding of how it improves physiological functions, mitigates disease risks, and establishes a theoretical foundation for employing physical exercise as a non-pharmacological intervention in disease prevention and treatment.
Collapse
Affiliation(s)
- Xi Zheng
- Fujian Key Laboratory of Developmental and Neural Biology & Southern Center for Biomedical Research, College of Life Sciences, Fujian Normal University, Fuzhou, PR China
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, PR China
| | - Xueli Liu
- Fujian Key Laboratory of Developmental and Neural Biology & Southern Center for Biomedical Research, College of Life Sciences, Fujian Normal University, Fuzhou, PR China
| | - Yuqian Guo
- Fujian Key Laboratory of Developmental and Neural Biology & Southern Center for Biomedical Research, College of Life Sciences, Fujian Normal University, Fuzhou, PR China
| | - Yi Lv
- Fujian Key Laboratory of Developmental and Neural Biology & Southern Center for Biomedical Research, College of Life Sciences, Fujian Normal University, Fuzhou, PR China
| | - Chensheng Lin
- Fujian Key Laboratory of Developmental and Neural Biology & Southern Center for Biomedical Research, College of Life Sciences, Fujian Normal University, Fuzhou, PR China
| | - Dan Wang
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, PR China
| | - Shaobing Wang
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, PR China
| | - Yiping Liu
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, PR China.
| | - Xuefeng Hu
- Fujian Key Laboratory of Developmental and Neural Biology & Southern Center for Biomedical Research, College of Life Sciences, Fujian Normal University, Fuzhou, PR China.
| |
Collapse
|
36
|
Liu H, Chen S, Xiang H, Xiao J, Zhao S, Zhang X, Shu Z, Zhang J, Ouyang J, Liu Q, Quan Q, Fan J, Gao P, Zheng X, Chen AF, Lu H. S1PR3 in hippocampal neurons improves synaptic plasticity and decreases depressive behavior via downregulation of RhoA/ROCK1. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111256. [PMID: 39828081 DOI: 10.1016/j.pnpbp.2025.111256] [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: 05/12/2024] [Revised: 10/30/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
The study investigates how Sphingosine-1-phosphate receptor 3 (S1PR3) and the Chronic Unpredictable Mild Stress (CUMS) affects depression-like behaviors. The S1P/S1PR3 signaling pathway is known to play a role in mood regulation, but it is not yet fully understood how it is connected to depression. This study looks to further explore this topic. To investigate the effect of CUMS on S1PR3 expression in hippocampus neurons and the synaptic plasticity, we observed animals' behavior with Sucrose Preference Test (SPT), Forced Swim Test (FST) and Open Field Test (OFT). Combining molecular and histological analysis, we investigated the S1PR3 expression, the change in synapse density, and synaptic structure change in the hippocampus. The CUMS caused a significant decrease in the S1PR3 expression, the density of the synaptic spine and synaptic ultrastructure change in mice. On the other hand, over-expression of S1PR3 by adeno-associated virus (AAV) in hippocampal neurons alleviated the depressive-like behaviors and synaptic deficits observed in stress-susceptible animals. Furthermore, the depressive-like phenotype and synaptic impairments were normalized by the expression of RhoA, implicating the RhoA/ROCK1 pathway in S1PR3 actions. Collectively, our findings provide strong evidence that S1PR3 plays a key role in hippocampal synaptic plasticity and depression and that modulation of S1PR3/RhoA/ROCK1 signaling may offer a novel therapeutic strategy for MDD. This study not only underscores the therapeutic potential of S1PR3 but also provides novel insights into the molecular mechanisms underlying depression.
Collapse
Affiliation(s)
- Huiqin Liu
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Shuhua Chen
- Department of Biochemistry, School of Life Sciences of Central South University, Changsha, China
| | - Hong Xiang
- Center for Experimental Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jie Xiao
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Shaoli Zhao
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xiao Zhang
- Department of Biochemistry, School of Life Sciences of Central South University, Changsha, China
| | - Zhihao Shu
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jing Zhang
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jie Ouyang
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Quanjun Liu
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Qisheng Quan
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jianing Fan
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Peng Gao
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xinru Zheng
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Alex F Chen
- Center for Experimental Medicine, The Third Xiangya Hospital of Central South University, Changsha, China; Institute for Cardiovascular Development and Regenerative Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hongwei Lu
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
37
|
Zeng CW. Stem Cell-Based Approaches for Spinal Cord Injury: The Promise of iPSCs. BIOLOGY 2025; 14:314. [PMID: 40136570 PMCID: PMC11940451 DOI: 10.3390/biology14030314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/09/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025]
Abstract
Spinal cord injury (SCI) is a life-altering condition that leads to severe neurological deficits and significantly impacts patients' quality of life. Despite advancements in medical care, current treatment options remain largely palliative, with limited ability to promote meaningful functional recovery. Induced pluripotent stem cells (iPSCs) have emerged as a promising avenue for regenerative medicine, offering patient-specific, cell-based therapeutic potential for SCI repair. This review provides a comprehensive overview of recent advancements in iPSC-based approaches for SCI, detailing the strategies used to generate neural cell types, including neural progenitor cells, oligodendrocytes, astrocytes, and microglia, and their roles in promoting neuroprotection and regeneration. Additionally, we examine key preclinical and clinical studies, highlighting functional recovery assessments and discussing both standardized and debated evaluation metrics. Furthermore, we address critical challenges related to safety, tumorigenicity, immune response, survival, integration, and overcoming the inhibitory microenvironment of the injured spinal cord. We also explore emerging approaches in biomaterial scaffolds, gene editing, and rehabilitation strategies that may enhance the clinical applicability of iPSC-based therapies. By addressing these challenges and refining translational strategies, iPSC-based interventions hold significant potential to revolutionize SCI treatment and improve outcomes for affected individuals.
Collapse
Affiliation(s)
- Chih-Wei Zeng
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
38
|
Belančić A, Janković T, Gkrinia EMM, Kristić I, Rajič Bumber J, Rački V, Pilipović K, Vitezić D, Mršić-Pelčić J. Glial Cells in Spinal Muscular Atrophy: Speculations on Non-Cell-Autonomous Mechanisms and Therapeutic Implications. Neurol Int 2025; 17:41. [PMID: 40137462 PMCID: PMC11944370 DOI: 10.3390/neurolint17030041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Spinal muscular atrophy (SMA) is a neuromuscular disorder caused by homozygous deletions or mutations in the SMN1 gene, leading to progressive motor neuron degeneration. While SMA has been classically viewed as a motor neuron-autonomous disease, increasing evidence indicates a significant role of glial cells-astrocytes, microglia, oligodendrocytes, and Schwann cells-in the disease pathophysiology. Astrocytic dysfunction contributes to motor neuron vulnerability through impaired calcium homeostasis, disrupted synaptic integrity, and neurotrophic factor deficits. Microglia, through reactive gliosis and complement-mediated synaptic stripping, exacerbate neurodegeneration and neuroinflammation. Oligodendrocytes exhibit impaired differentiation and metabolic support, while Schwann cells display abnormalities in myelination, extracellular matrix composition, and neuromuscular junction maintenance, further compromising motor function. Dysregulation of pathways such as NF-κB, Notch, and JAK/STAT, alongside the upregulation of complement proteins and microRNAs, reinforces the non-cell-autonomous nature of SMA. Despite the advances in SMN-restorative therapies, they do not fully mitigate glial dysfunction. Targeting glial pathology, including modulation of reactive astrogliosis, microglial polarization, and myelination deficits, represents a critical avenue for therapeutic intervention. This review comprehensively examines the multifaceted roles of glial cells in SMA and highlights emerging glia-targeted strategies to enhance treatment efficacy and improve patient outcomes.
Collapse
Affiliation(s)
- Andrej Belančić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Tamara Janković
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | | | - Iva Kristić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Jelena Rajič Bumber
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Valentino Rački
- Department of Neurology, Clinical Hospital Centre Rijeka, Krešimirova 42, 51000 Rijeka, Croatia;
| | - Kristina Pilipović
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Dinko Vitezić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Jasenka Mršić-Pelčić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| |
Collapse
|
39
|
Antal M. Molecular Anatomy of Synaptic and Extrasynaptic Neurotransmission Between Nociceptive Primary Afferents and Spinal Dorsal Horn Neurons. Int J Mol Sci 2025; 26:2356. [PMID: 40076973 PMCID: PMC11900602 DOI: 10.3390/ijms26052356] [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: 12/04/2024] [Revised: 02/26/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Sensory signals generated by peripheral nociceptors are transmitted by peptidergic and nonpeptidergic nociceptive primary afferents to the superficial spinal dorsal horn, where their central axon terminals establish synaptic contacts with secondary sensory spinal neurons. In the case of suprathreshold activation, the axon terminals release glutamate into the synaptic cleft and stimulate postsynaptic spinal neurons by activating glutamate receptors located on the postsynaptic membrane. When overexcitation is evoked by peripheral inflammation, neuropathy or pruritogens, peptidergic nociceptive axon terminals may corelease various neuropeptides, neurotrophins and endomorphin, together with glutamate. However, in contrast to glutamate, neuropeptides, neurotrophins and endomorphin are released extrasynaptically. They diffuse from the site of release and modulate the function of spinal neurons via volume transmission, activating specific extrasynaptic receptors. Thus, the released neuropeptides, neurotrophins and endomorphin may evoke excitation, disinhibition or inhibition in various spinal neuronal populations, and together with glutamate, induce overall overexcitation, called central sensitization. In addition, the synaptic and extrasynaptic release of neurotransmitters is subjected to strong retrograde control mediated by various retrogradely acting transmitters, messengers, and their presynaptic receptors. Moreover, the composition of this complex chemical apparatus is heavily dependent on the actual patterns of nociceptive primary afferent activation in the periphery. This review provides an overview of the complexity of this signaling apparatus, how nociceptive primary afferents can activate secondary sensory spinal neurons via synaptic and volume transmission in the superficial spinal dorsal horn, and how these events can be controlled by presynaptic mechanisms.
Collapse
Affiliation(s)
- Miklós Antal
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| |
Collapse
|
40
|
Xu S, Jiang X, Xu M, Ai C, Zhao G, Jiang T, Liu Y, Tian Z, Zhang M, Dong J. Design, synthesis and biological evaluation of novel 1H-indole-3-carbonitrile derivatives as potent TRK Inhibitors. Eur J Med Chem 2025; 285:117231. [PMID: 39778327 DOI: 10.1016/j.ejmech.2024.117231] [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: 11/17/2024] [Revised: 12/17/2024] [Accepted: 12/29/2024] [Indexed: 01/11/2025]
Abstract
Tropomyosin receptor kinase (TRK) has emerged as a promising therapeutic target in cancers driven by NTRK gene fusions. Herein, we report a highly potent TRK inhibitor, C11, developed using bioisosteric replacement and computer-aided drug design (CADD) strategies. Compound C11 demonstrated significant antiproliferative effects against TRK-dependent cell lines (Km-12), and exhibited a dose-dependent inhibition of both colony formation and cell migration. Mechanistic study revealed that C11 induced cancer cell death by arresting the cell cycle, triggering apoptosis, and reducing phosphorylated TRK levels. In vitro stability assays showed that compound C11 possessed excellent plasma stability (t1/2 > 480 min) and moderate liver microsomal stability (t1/2 = 38.9 min). Pharmacokinetic evaluation further indicated an oral bioavailability of 15.2 % for compound C11. These results highlight compound C11 as a promising lead compound for the further development of TRK inhibitors.
Collapse
Affiliation(s)
- Shaoshan Xu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xiaosheng Jiang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China; Department of Chemistry and Biochemistry, Warren Family Research Center for Drug Discovery and Development, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Mengdi Xu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Chengjian Ai
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Guanyi Zhao
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Tao Jiang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yang Liu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Zhen Tian
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Meihui Zhang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Jinhua Dong
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
41
|
Kim S, Park J, Eo H, Lee GB, Park SM, Shin M, Lee SE, Nam Y, Kim SR. Intracerebellar upregulation of Rheb(S16H) ameliorates motor dysfunction in mice with SCA2. Acta Pharmacol Sin 2025:10.1038/s41401-025-01504-y. [PMID: 40033054 DOI: 10.1038/s41401-025-01504-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: 10/15/2024] [Accepted: 02/04/2025] [Indexed: 03/05/2025]
Abstract
Cerebellar ataxia (CA) is characterized by impaired balance and coordination due to the loss of cerebellar neurons caused by various factors, and effective treatments are currently lacking. Recently, we observed reduced expression of signaling molecules in the mammalian target of rapamycin complex 1 (mTORC1) pathway in the cerebellum of mice with spinocerebellar ataxia type 2 (SCA2) compared with wild-type mice. To investigate the effects of mTORC1 upregulation on motor dysfunction in mice with SCA2, we administered an intracerebellar injection of adeno-associated virus serotype 1 carrying a constitutively active form of Ras homolog enriched in brain [Rheb(S16H)], which is an upstream activator of mTORC1. This treatment led to increased Rheb(S16H) expression in calbindin-D28K-positive Purkinje cells and increased levels of neurotrophic factors. Additionally, Rheb(S16H) upregulation reduced abnormal behaviors and protected Purkinje cells in mice with SCA2. Our findings suggest that upregulating Rheb(S16H) in the cerebellum may be a promising therapeutic strategy for hereditary CA.
Collapse
Affiliation(s)
- Sehwan Kim
- School of Life Science and Biotechnology, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41404, Republic of Korea
| | - Junwoo Park
- School of Life Science and Biotechnology, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Hyemi Eo
- School of Life Science and Biotechnology, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Gi Beom Lee
- School of Life Science and Biotechnology, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Se Min Park
- School of Life Science and Biotechnology, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Minsang Shin
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41404, Republic of Korea
- Department of Microbiology, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Seung Eun Lee
- Research Animal Resource Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Youngpyo Nam
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41404, Republic of Korea.
| | - Sang Ryong Kim
- School of Life Science and Biotechnology, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Republic of Korea.
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41404, Republic of Korea.
| |
Collapse
|
42
|
Mucci F, Arone A, Gurrieri R, Weiss F, Russomanno G, Marazziti D. Third-Generation Antipsychotics: The Quest for the Key to Neurotrophism. Life (Basel) 2025; 15:391. [PMID: 40141736 PMCID: PMC11944073 DOI: 10.3390/life15030391] [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: 12/27/2024] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Antipsychotic drugs (APs) have profoundly changed the treatment landscape for psychiatric disorders, yet their impact on neuroplasticity and neurotrophism remains only partially understood. While second-generation antipsychotics (SGAs) are associated with a better side effect profile than their predecessors, the emergence of third-generation antipsychotics (TGAs)-such as brexpiprazole, cariprazine, lurasidone, iloperidone, lumateperone, pimavanserin, and roluperidone-has prompted renewed interest in their potential neuroprotective and pro-cognitive effects. This review attempts to carefully examine the evidence on the neurotrophic properties of TGAs and their role in modulating brain plasticity by analyzing studies published between 2010 and 2024. Although data remain limited and focused primarily on earlier SGAs, emerging findings suggest that some TGAs may exert positive effects on neuroplastic processes, including the modulation of brain-derived neurotrophic factors (BDNFs) and synaptic architecture. However, robust clinical data on their long-term effects and comparative efficacy are lacking; therefore, further research is necessary to validate their role in preventing neurodegenerative changes and improving cognitive outcomes in patients with psychiatric conditions.
Collapse
Affiliation(s)
- Federico Mucci
- Department of Psychiatry, Lucca Zone, Azienda USL Toscana Nord Ovest, 55100 Lucca, Italy;
| | - Alessandro Arone
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, 56100 Pisa, Italy; (A.A.); (R.G.); (F.W.); (G.R.)
| | - Riccardo Gurrieri
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, 56100 Pisa, Italy; (A.A.); (R.G.); (F.W.); (G.R.)
| | - Francesco Weiss
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, 56100 Pisa, Italy; (A.A.); (R.G.); (F.W.); (G.R.)
| | - Gerardo Russomanno
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, 56100 Pisa, Italy; (A.A.); (R.G.); (F.W.); (G.R.)
| | - Donatella Marazziti
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, 56100 Pisa, Italy; (A.A.); (R.G.); (F.W.); (G.R.)
| |
Collapse
|
43
|
He Y, Ye M, Xia Y, Zhong Z, Wang W, Li Q. The role of cytokines as predictors for NAFLD-related diseases: A bidirectional Mendelian randomization study. Clin Res Hepatol Gastroenterol 2025; 49:102545. [PMID: 39900199 DOI: 10.1016/j.clinre.2025.102545] [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: 11/28/2024] [Revised: 01/26/2025] [Accepted: 01/31/2025] [Indexed: 02/05/2025]
Abstract
BACKGROUND Prior research has highlighted associations between inflammatory cytokines and non-alcoholic fatty liver disease (NAFLD), but causal relationships remain unclear. Employing the Mendelian randomization (MR) approach, this investigation aims to explore the connection between 41 inflammatory cytokines and NAFLD-related diseases. METHODS Our research implemented bidirectional study focusing on 41 cytokines in 8,293 Finns, predicting genetic associations with NAFLD, nonalcoholic steatohepatitis (NASH), fibrosis, and cirrhosis. We primarily utilized the inverse variance weighted (IVW) method to evaluate the bidirectional relationships. Additionally, a sensitivity analysis was carried out to ensure the reliability of our findings. RESULTS An elevated risk for NAFLD was correlated with both IL-2 (OR = 1.226, 95 % CI = 1.018-1.477, p = 0.031) and TNF-β (OR = 1.151, 95 % CI = 1.011-1.310, p = 0.033). IL-16 is associated with decreased NAFLD risk (OR = 0.820, 95 % CI = 0.719-0.934, p = 0.033). β-NGF (OR = 2.495, 95 % CI = 1.019-6.108, p = 0.045) and SCGFβ (OR = 1.541, 95 % CI = 1.052-2.256, p = 0.026) are linked to higher NASH risk. No significant associations were found for fibrosis and cirrhosis. Furthermore, the causal relationship between genetic predisposition to NAFLD-related diseases and various inflammatory cytokines was established. CONCLUSIONS Our MR analysis identifies specific cytokines as genetic predictors for NAFLD and NASH. IL-2 and TNF-β increase NAFLD risk, IL-16 appears protective, and β-NGF and SCGFβ are associated with greater NASH risk. These insights are crucial for understanding the etiology and treatment of NAFLD-related diseases.
Collapse
Affiliation(s)
- Yijia He
- Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Miaomin Ye
- Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yin Xia
- Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ziyi Zhong
- Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Weiping Wang
- Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qian Li
- Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
44
|
Katz DH, Lindholm ME, Ashley EA. Charting the Molecular Terrain of Exercise: Energetics, Exerkines, and the Future of Multiomic Mapping. Physiology (Bethesda) 2025; 40:0. [PMID: 39136551 DOI: 10.1152/physiol.00024.2024] [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: 05/07/2024] [Revised: 08/09/2024] [Accepted: 08/09/2024] [Indexed: 11/21/2024] Open
Abstract
Physical activity plays a fundamental role in human health and disease. Exercise has been shown to improve a wide variety of disease states, and the scientific community is committed to understanding the precise molecular mechanisms that underlie the exquisite benefits. This review provides an overview of molecular responses to acute exercise and chronic training, particularly energy mobilization and generation, structural adaptation, inflammation, and immune regulation. Furthermore, it offers a detailed discussion of known molecular signals and systemic regulators activated during various forms of exercise and their role in orchestrating health benefits. Critically, the increasing use of multiomic technologies is explored with an emphasis on how multiomic and multitissue studies contribute to a more profound understanding of exercise biology. These data inform anticipated future advancement in the field and highlight the prospect of integrating exercise with pharmacology for personalized disease prevention and treatment.
Collapse
Affiliation(s)
- Daniel H Katz
- Division of Cardiovascular MedicineStanford University School of Medicine, Stanford, California, United States
| | - Maléne E Lindholm
- Division of Cardiovascular MedicineStanford University School of Medicine, Stanford, California, United States
| | - Euan A Ashley
- Division of Cardiovascular MedicineStanford University School of Medicine, Stanford, California, United States
| |
Collapse
|
45
|
Ma L, Zhang Z, Mu Y, Liu B, Zhou H, Wang DA. The Application of Biomaterial-Based Spinal Cord Tissue Engineering. Macromol Biosci 2025; 25:e2400444. [PMID: 39472074 DOI: 10.1002/mabi.202400444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/11/2024] [Indexed: 03/14/2025]
Abstract
Advancements in biomaterial-based spinal cord tissue engineering technology have profoundly influenced regenerative medicine, providing innovative solutions for both spinal cord organoid development and engineered spinal cord injury (SCI) repair. In spinal cord organoids, biomaterials offer a supportive microenvironment that mimics the natural extracellular matrix, facilitating cell differentiation and organization and advancing the understanding of spinal cord development and pathophysiology. Furthermore, biomaterials are essential in constructing engineered spinal cords for SCI repair. The incorporation of biomaterials with growth factors, fabrication of ordered scaffold structures, and artificial spinal cord assemblies are critical insights for SCI to ensure structural integrity, enhance cell viability, and promote neural regeneration in transplantation. In summary, this review summarizes the contribution of biomaterials to the spinal cord organoids progression and discusses strategies for biomaterial-based spinal cord engineering in SCI therapy. These achievements underscore the transformative potential of biomaterials to improve treatment options for SCI and accelerate future clinical applications.
Collapse
Affiliation(s)
- Liang Ma
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
| | - Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
| | - Yulei Mu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
| | - Bangheng Liu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
| | - Huiqun Zhou
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
- Centre for Neuromusculoskeletal Restorative Medicine, InnoHK HKSTP, Sha Tin, Hong Kong, 999077, China
- Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
| |
Collapse
|
46
|
Puhlmann LM, Vrtička P, Linz R, Valk SL, Papassotiriou I, Chrousos GP, Engert V, Singer T. Serum BDNF Increase After 9-Month Contemplative Mental Training Is Associated With Decreased Cortisol Secretion and Increased Dentate Gyrus Volume: Evidence From a Randomized Clinical Trial. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100414. [PMID: 39896238 PMCID: PMC11786774 DOI: 10.1016/j.bpsgos.2024.100414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 10/06/2024] [Accepted: 11/04/2024] [Indexed: 02/04/2025] Open
Abstract
Background In this study, we investigated whether mindfulness- and meditation-based mental training that improves stress regulation can upregulate BDNF (brain-derived neurotrophic factor), an important promoter of hippocampal neuroplasticity, and examined cortisol reduction as a mediating pathway. Methods In a randomized clinical trial, 332 healthy adults were randomly assigned to one of the 3 training cohorts or a passive control cohort. Training participants completed up to three 3-month-long modules targeting attention-based mindfulness, socio-affective skills, or socio-cognitive skills. We examined change in serum BDNF levels after each 3-month training interval; evaluated whether training effects were linked to reduced cortisol release in the long-term, diurnally, and when acutely stress-induced; and explored associations with hippocampal volume changes. Results In the combined training cohorts, BDNF increased significantly and cumulatively after 3-, 6-, and 9-month training relative to the pretraining baseline (3 month: t 516 = 3.57 [estimated increase: 1353 pg/mL], 6 month: t 516 = 3.45 [1557 pg/mL], 9 month: t 516 = 3.45 [2276 pg/mL]; all ps < .001). After 9 months, training cohort BDNF was not higher than control cohort BDNF, which displayed unexplained variance. However, moderated mediation analysis showed that only training effects, and not control cohort BDNF change, were partially mediated by simultaneously reduced long-term cortisol release (3-month averages) measured in hair (15.1% mediation, p = .021). Individually greater BDNF increase after training correlated with more reduced long-term and stress-induced cortisol release. Moreover, greater BDNF increase after 9 months of training correlated with dentate gyrus volume increase (t 108 = 2.09, p = .039). Conclusions Longitudinal contemplative training may promote a neurobiological pathway from stress reduction to increased BDNF levels to enhanced hippocampal volume. However, single serum BDNF measurements can be unreliable for assessing long-term neurotrophic effects in healthy adults. Future studies should investigate nonspecific BDNF measurement effects before considering application in preventive health care.
Collapse
Affiliation(s)
- Lara M.C. Puhlmann
- Research Group Social Stress and Family Health, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Leibniz Institute for Resilience Research, Mainz, Germany
| | - Pascal Vrtička
- Research Group Social Stress and Family Health, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Centre for Brain Science, Department of Psychology, University of Essex, Colchester, United Kingdom
| | - Roman Linz
- Research Group Social Stress and Family Health, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Sofie L. Valk
- Otto Hahn Group Cognitive Neurogenetics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Institute of Neuroscience and Medicine (INM-7: Brain & Behaviour), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Ioannis Papassotiriou
- Department of Clinical Biochemistry, Aghia Sophia Children’s Hospital, Athens, Greece
| | - George P. Chrousos
- First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, Aghia Sophia Children’s Hospital, Athens, Greece
| | - Veronika Engert
- Research Group Social Stress and Family Health, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Institute for Psychosocial Medicine, Psychotherapy and Psychooncology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
- German Center for Mental Health, partner site Halle-Jena-Magdeburg, Jena, Germany
- Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying Mental Health, Halle-Jena-Magdeburg, Jena, Germany
| | - Tania Singer
- Social Neuroscience Laboratory, Max Planck Society, Berlin, Germany
| |
Collapse
|
47
|
Arslankiran A, Acikgoz B, Demirtas H, Dalkiran B, Kiray A, Aksu I, Kiray M, Dayi A. Effects of voluntary or involuntary exercise in adolescent male rats exposed to chronic social isolation on cognition, behavior, and neurotrophic factors. Biol Futur 2025; 76:71-85. [PMID: 39966302 DOI: 10.1007/s42977-025-00250-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/09/2025] [Indexed: 02/20/2025]
Abstract
This study investigated the effects of voluntary and involuntary/regular exercise on neurotrophic factors in the brain, cognitive functions, and anxiety in socially isolated adolescent male rats. In this study, 42 adolescent male Sprague-Dawley rats were divided into six groups: control (C), socially isolated (SI), voluntary exercise (VE), regular exercise (RE), socially isolated + voluntary exercise (SI-VE), and socially isolated + regular exercise (SI-RE). Socially isolated groups were kept in separate cages for 4 weeks. Treadmill and wheel running were used in the exercise groups. The following behavioral tests-elevated plus maze (EPM), open field, ultrasonic vocalization (USV), and Morris water maze (MWM)-rats were euthanized, and brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), and vascular endothelial growth factor (VEGF) levels were measured in the prefrontal cortex (PFC) and hippocampus. Statistically, the differences between the groups were evaluated by one-way ANOVA and post hoc LSD tests using IBM SPSS software. In the EPM, locomotor activity was higher in the voluntary exercise groups. In the MWM, both regular exercise groups found the platform faster. In the USV, the SI-RE group produced more 50-kHz sounds. BDNF and NGF levels in the hippocampus were higher in the SI-RE and SI-VE groups; VEGF levels were higher in the SI-RE group. Neuron density in the PFC increased in the SI-RE and VE groups, while neuron density in the hippocampus increased in the SI-RE, SI-VE, and VE groups. According to the findings, we showed that voluntary exercise reduces social isolation-induced anxiety, and involuntary/regular exercise both reduces anxiety and has potential benefits on cognitive functions.
Collapse
Affiliation(s)
- Ayca Arslankiran
- Department of Physiology, School of Medicine, Dokuz Eylul University, Izmir, Turkey.
- Institute of Health Sciences, Dokuz Eylul University, Izmir, Turkey.
| | - Burcu Acikgoz
- Department of Physiology, School of Medicine, Dokuz Eylul University, Izmir, Turkey
- Institute of Health Sciences, Dokuz Eylul University, Izmir, Turkey
| | - Helin Demirtas
- Department of Physiology, School of Medicine, Dokuz Eylul University, Izmir, Turkey
- Institute of Health Sciences, Dokuz Eylul University, Izmir, Turkey
| | - Bahar Dalkiran
- Department of Physiology, School of Medicine, Dokuz Eylul University, Izmir, Turkey
- Institute of Health Sciences, Dokuz Eylul University, Izmir, Turkey
| | - Amac Kiray
- Department of Anatomy, School of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Ilkay Aksu
- Department of Physiology, School of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Muge Kiray
- Department of Physiology, School of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Ayfer Dayi
- Department of Physiology, School of Medicine, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
48
|
Chin SE, Gallego P, Aagaard A, Carmen S, Barrett N, Wolny M, Cloarec S, Paterson J, Sivapalan R, Hunt J, Murray TV, Delaney T, Sjögren T, Neal F. Identification of unique binding mode anti-NTF3 antibodies from a novel long VH CDR3 phage display library. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2025; 31:100216. [PMID: 39832740 DOI: 10.1016/j.slasd.2025.100216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Neurotrophic factor 3 (NTF3) is a cysteine knot protein and a member of the nerve growth factor (NGF) family of cytokines. NTF3 engages the Trk family of receptor tyrosine kinases, playing a pivotal role in the development and function of both the central and peripheral nervous systems. Its involvement in neuronal survival, differentiation, and growth links NTF3 to a spectrum of neurodegenerative diseases. Consequently, targeting NTF3 with antibodies holds promise as a first in class therapeutic opportunity for a wide range of conditions. Specific and neutralizing antibodies against NTF3 were successfully isolated using phage display. Initial phage display selections revealed a preference of hits for a longer than average complementarity-determining region 3 (CDR3) in the heavy chain variable domain (VH). To investigate this further we developed a long loop length VH CDR3 antibody library that demonstrated increased hit rates versus a standard antibody library and allowed the isolation of IgG that demonstrated inhibition of functional activity, coupled with a favourable kinetic profile. Structural analysis of the Fab/NTF3 interaction, via X-ray crystallography, unveiled an unconventional interaction wherein regions beyond the longer CDR loops of the Fab induced ordering in a flexible loop on NTF3, which remained disordered in its free antigenic state. This comprehensive approach not only sheds light on the therapeutic potential of NTF3-specific antibodies but also provides critical structural details that enhance our understanding of the complex NTF3-Fab interaction thus offering valuable insights for future antibody design and therapeutic development.
Collapse
Affiliation(s)
- Stacey E Chin
- Biologics Engineering, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Pablo Gallego
- Structure and BioPhysics, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anna Aagaard
- Structure and BioPhysics, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Sara Carmen
- Former AstraZeneca employee, Biologics Engineering, Oncology R&D, AstraZeneca, Cambridge, UK
| | | | - Marcin Wolny
- Biologics Engineering, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Sophie Cloarec
- Biologics Engineering, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Judy Paterson
- Biologics Engineering, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Rohan Sivapalan
- Biologics Engineering, Oncology R&D, AstraZeneca, Cambridge, UK
| | - James Hunt
- Biologics Engineering, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Thomas V Murray
- Biologics Engineering, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Tracy Delaney
- Bioscience COPD/IPF, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | - Tove Sjögren
- Structure and BioPhysics, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Frances Neal
- Biologics Engineering, Oncology R&D, AstraZeneca, Cambridge, UK.
| |
Collapse
|
49
|
Zhang J, Kwan HLR, Chan CB, Lee CW. Localized release of muscle-generated BDNF regulates the initial formation of postsynaptic apparatus at neuromuscular synapses. Cell Death Differ 2025; 32:546-560. [PMID: 39511403 PMCID: PMC11893767 DOI: 10.1038/s41418-024-01404-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/14/2024] [Accepted: 10/18/2024] [Indexed: 11/15/2024] Open
Abstract
Growing evidence indicates that brain-derived neurotrophic factor (BDNF) is produced in contracting skeletal muscles and is secreted as a myokine that plays an important role in muscle metabolism. However, the involvement of muscle-generated BDNF and the regulation of its vesicular trafficking, localization, proteolytic processing, and spatially restricted release during the development of vertebrate neuromuscular junctions (NMJs) remain largely unknown. In this study, we first reported that BDNF is spatially associated with the actin-rich core domain of podosome-like structures (PLSs) at topologically complex acetylcholine receptor (AChR) clusters in cultured Xenopus muscle cells. The release of spatially localized BDNF is tightly controlled by activity-regulated mechanisms in a calcium-dependent manner. Live-cell time-lapse imaging further showed that BDNF-containing vesicles are transported to and captured at PLSs in both aneural and synaptic AChR clusters for spatially restricted release. Functionally, BDNF knockdown or furin-mediated endoproteolytic activity inhibition significantly suppresses aneural AChR cluster formation, which in turn affects synaptic AChR clustering induced by nerve innervation or agrin-coated beads. Lastly, skeletal muscle-specific BDNF knockout (MBKO) mice exhibit structural defects in the formation of aneural AChR clusters and their subsequent recruitment to nerve-induced synaptic AChR clusters during the initial stages of NMJ development in vivo. Together, this study demonstrated the regulatory roles of PLSs in the intracellular trafficking, spatial localization, and activity-dependent release of BDNF in muscle cells and revealed the involvement of muscle-generated BDNF and its proteolytic conversion in regulating the initial formation of aneural and synaptic AChR clusters during early NMJ development in vitro and in vivo.
Collapse
Affiliation(s)
- Jinkai Zhang
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Hong Kong, China
| | - Hiu-Lam Rachel Kwan
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chi Bun Chan
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong, China
| | - Chi Wai Lee
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Hong Kong, China.
- Golden Meditech Centre for NeuroRegeneration Sciences, Hong Kong Baptist University, Hong Kong, China.
| |
Collapse
|
50
|
Zanardo E, Quinto G, Battista F, Duregon F, Vecchiato M, Bergia C, Erickson K, Ermolao A, Neunhaeuserer D. Acute effects of physical exercise on cognitive function and neurotrophins in patients with type 1 diabetes: A systematic review. Heliyon 2025; 11:e42456. [PMID: 40028553 PMCID: PMC11868937 DOI: 10.1016/j.heliyon.2025.e42456] [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: 04/15/2024] [Revised: 02/01/2025] [Accepted: 02/03/2025] [Indexed: 03/05/2025] Open
Abstract
Background Type 1 diabetes mellitus (T1DM) is associated with cognitive decline. In contrast, higher levels of neurotrophins, such as brain-derived neurotrophic factor (BDNF), may be associated with better brain health. Physical exercise has been associated with elevated levels of BDNF and consequently improved cognitive function, but whether this association is found in T1DM remains unresolved. The aim of this systematic review was to evaluate the acute effect of physical exercise on cognitive function and BDNF levels in patients affected by T1DM. Methods MEDLINE, Cochrane Library (CENTRAL database), EMBASE and SPORTDiscus were screened by 2 independent reviewers, who selected studies that analysed acute effects of physical exercise in patients with T1DM on BDNF levels or cognitive function tests before and after exercise. Studies in humans and English written were included. The quality of these studies was assessed using the respective Cochrane Risk of Bias tool. Results After identifying 507 articles, 4 studies including 78 participants were analysed. Two studies were non-randomized clinical trials, the others were crossover trials. Selected studies performed different exercise intervention protocols, evaluating both high and moderate intensity training. BDNF levels were found higher after exercise in all studies. Cognitive function tests resulted also improved after the training intervention. Conclusions In subjects with T1DM, preliminary evidence suggests that exercise training might increase plasma BDNF levels and ameliorate cognitive deficits. However, scientific evidence is still very limited and there is a significant need for further research to clarify the possible positive neurocognitive effects of exercise in T1DM.
Collapse
Affiliation(s)
- Emanuele Zanardo
- Sport and Exercise Medicine Division, Department of Medicine, University of Padova, Padova, Italy
| | - Giulia Quinto
- Sport and Exercise Medicine Division, Department of Medicine, University of Padova, Padova, Italy
| | - Francesca Battista
- Sport and Exercise Medicine Division, Department of Medicine, University of Padova, Padova, Italy
| | - Federica Duregon
- Sport and Exercise Medicine Division, Department of Medicine, University of Padova, Padova, Italy
| | - Marco Vecchiato
- Sport and Exercise Medicine Division, Department of Medicine, University of Padova, Padova, Italy
| | - Chiara Bergia
- Sport and Exercise Medicine Division, Department of Medicine, University of Padova, Padova, Italy
| | - Kirk Erickson
- Advent Health Research Institute, Neuroscience, Orlando, FL, USA
- University of Pittsburgh, Department of Psychology, Pittsburgh, PA, USA
| | - Andrea Ermolao
- Sport and Exercise Medicine Division, Department of Medicine, University of Padova, Padova, Italy
| | - Daniel Neunhaeuserer
- Sport and Exercise Medicine Division, Department of Medicine, University of Padova, Padova, Italy
| |
Collapse
|