101
|
Chung J, Pierce J, Franklin C, Olson RM, Morrison AR, Amos-Landgraf J. Translating animal models of SARS-CoV-2 infection to vascular, neurological and gastrointestinal manifestations of COVID-19. Dis Model Mech 2025; 18:dmm052086. [PMID: 40195851 PMCID: PMC12010913 DOI: 10.1242/dmm.052086] [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: 04/09/2025] Open
Abstract
Since the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) initiated a global pandemic resulting in an estimated 775 million infections with over 7 million deaths, it has become evident that COVID-19 is not solely a pulmonary disease. Emerging evidence has shown that, in a subset of patients, certain symptoms - including chest pain, stroke, anosmia, dysgeusia, diarrhea and abdominal pain - all indicate a role of vascular, neurological and gastrointestinal (GI) pathology in the disease process. Many of these disease processes persist long after the acute disease has been resolved, resulting in 'long COVID' or post-acute sequelae of COVID-19 (PASC). The molecular mechanisms underlying the acute and systemic conditions associated with COVID-19 remain incompletely defined. Appropriate animal models provide a method of understanding underlying disease mechanisms at the system level through the study of disease progression, tissue pathology, immune system response to the pathogen and behavioral responses. However, very few studies have addressed PASC and whether existing models hold promise for studying this challenging problem. Here, we review the current literature on cardiovascular, neurological and GI pathobiology caused by COVID-19 in patients, along with established animal models of the acute disease manifestations and their prospects for use in PASC studies. Our aim is to provide guidance for the selection of appropriate models in order to recapitulate certain aspects of the disease to enhance the translatability of mechanistic studies.
Collapse
Affiliation(s)
- James Chung
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Julia Pierce
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Department of Research, Ocean State Research Institute, Inc., Providence, RI 02908-4734, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI 02908, USA
| | - Craig Franklin
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Rachel M. Olson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
- Laboratory for Infectious Disease Research, University of Missouri, Columbia, MO 65211, USA
| | - Alan R. Morrison
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Department of Research, Ocean State Research Institute, Inc., Providence, RI 02908-4734, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI 02908, USA
| | - James Amos-Landgraf
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
102
|
Qin Y, Zhu W, Guo T, Zhang Y, Xing T, Yin P, Li S, Li XJ, Yang S. Reduced mesencephalic astrocyte-derived neurotrophic factor expression by mutant androgen receptor contributes to neurodegeneration in a model of spinal and bulbar muscular atrophy pathology. Neural Regen Res 2025; 20:2655-2666. [PMID: 38934406 PMCID: PMC11801304 DOI: 10.4103/nrr.nrr-d-23-01666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 01/08/2024] [Accepted: 02/08/2024] [Indexed: 06/28/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202509000-00027/figure1/v/2024-11-05T132919Z/r/image-tiff Spinal and bulbar muscular atrophy is a neurodegenerative disease caused by extended CAG trinucleotide repeats in the androgen receptor gene, which encodes a ligand-dependent transcription factor. The mutant androgen receptor protein, characterized by polyglutamine expansion, is prone to misfolding and forms aggregates in both the nucleus and cytoplasm in the brain in spinal and bulbar muscular atrophy patients. These aggregates alter protein-protein interactions and compromise transcriptional activity. In this study, we reported that in both cultured N2a cells and mouse brain, mutant androgen receptor with polyglutamine expansion causes reduced expression of mesencephalic astrocyte-derived neurotrophic factor. Overexpression of mesencephalic astrocyte-derived neurotrophic factor ameliorated the neurotoxicity of mutant androgen receptor through the inhibition of mutant androgen receptor aggregation. Conversely, knocking down endogenous mesencephalic astrocyte-derived neurotrophic factor in the mouse brain exacerbated neuronal damage and mutant androgen receptor aggregation. Our findings suggest that inhibition of mesencephalic astrocyte-derived neurotrophic factor expression by mutant androgen receptor is a potential mechanism underlying neurodegeneration in spinal and bulbar muscular atrophy.
Collapse
Affiliation(s)
- Yiyang Qin
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong–HongKong–Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Wenzhen Zhu
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong–HongKong–Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Tingting Guo
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong–HongKong–Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Yiran Zhang
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong–HongKong–Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Tingting Xing
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong–HongKong–Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Peng Yin
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong–HongKong–Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Shihua Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong–HongKong–Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong–HongKong–Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Su Yang
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong–HongKong–Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| |
Collapse
|
103
|
Su C, Huang T, Zhang M, Zhang Y, Zeng Y, Chen X. Glucocorticoid receptor signaling in the brain and its involvement in cognitive function. Neural Regen Res 2025; 20:2520-2537. [PMID: 39248182 PMCID: PMC11801288 DOI: 10.4103/nrr.nrr-d-24-00355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/07/2024] [Accepted: 07/06/2024] [Indexed: 09/10/2024] Open
Abstract
The hypothalamic-pituitary-adrenal axis regulates the secretion of glucocorticoids in response to environmental challenges. In the brain, a nuclear receptor transcription factor, the glucocorticoid receptor, is an important component of the hypothalamic-pituitary-adrenal axis's negative feedback loop and plays a key role in regulating cognitive equilibrium and neuroplasticity. The glucocorticoid receptor influences cognitive processes, including glutamate neurotransmission, calcium signaling, and the activation of brain-derived neurotrophic factor-mediated pathways, through a combination of genomic and non-genomic mechanisms. Protein interactions within the central nervous system can alter the expression and activity of the glucocorticoid receptor, thereby affecting the hypothalamic-pituitary-adrenal axis and stress-related cognitive functions. An appropriate level of glucocorticoid receptor expression can improve cognitive function, while excessive glucocorticoid receptors or long-term exposure to glucocorticoids may lead to cognitive impairment. Patients with cognitive impairment-associated diseases, such as Alzheimer's disease, aging, depression, Parkinson's disease, Huntington's disease, stroke, and addiction, often present with dysregulation of the hypothalamic-pituitary-adrenal axis and glucocorticoid receptor expression. This review provides a comprehensive overview of the functions of the glucocorticoid receptor in the hypothalamic-pituitary-adrenal axis and cognitive activities. It emphasizes that appropriate glucocorticoid receptor signaling facilitates learning and memory, while its dysregulation can lead to cognitive impairment. This provides clues about how glucocorticoid receptor signaling can be targeted to overcome cognitive disability-related disorders.
Collapse
Affiliation(s)
- Chonglin Su
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Taiqi Huang
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Meiyu Zhang
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Yanyu Zhang
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Yan Zeng
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Xingxing Chen
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
104
|
Long Z, Ge C, Zhao Y, Liu Y, Zeng Q, Tang Q, Dong Z, He G. Enhanced autophagic clearance of amyloid-β via histone deacetylase 6-mediated V-ATPase assembly and lysosomal acidification protects against Alzheimer's disease in vitro and in vivo. Neural Regen Res 2025; 20:2633-2644. [PMID: 38993141 PMCID: PMC11801289 DOI: 10.4103/nrr.nrr-d-23-01633] [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: 09/28/2023] [Revised: 02/05/2024] [Accepted: 03/29/2024] [Indexed: 07/13/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202509000-00025/figure1/v/2024-11-05T132919Z/r/image-tiff Recent studies have suggested that abnormal acidification of lysosomes induces autophagic accumulation of amyloid-β in neurons, which is a key step in senile plaque formation. Therefore, restoring normal lysosomal function and rebalancing lysosomal acidification in neurons in the brain may be a new treatment strategy for Alzheimer's disease. Microtubule acetylation/deacetylation plays a central role in lysosomal acidification. Here, we show that inhibiting the classic microtubule deacetylase histone deacetylase 6 with an histone deacetylase 6 shRNA or thehistone deacetylase 6 inhibitor valproic acid promoted lysosomal reacidification by modulating V-ATPase assembly in Alzheimer's disease. Furthermore, we found that treatment with valproic acid markedly enhanced autophagy, promoted clearance of amyloid-β aggregates, and ameliorated cognitive deficits in a mouse model of Alzheimer's disease. Our findings demonstrate a previously unknown neuroprotective mechanism in Alzheimer's disease, in which histone deacetylase 6 inhibition by valproic acid increases V-ATPase assembly and lysosomal acidification.
Collapse
Affiliation(s)
- Zhimin Long
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Chuanhua Ge
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Yueyang Zhao
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Yuanjie Liu
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Qinghua Zeng
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Qing Tang
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Physiology, Chongqing Medical University, Chongqing, China
| | - Zhifang Dong
- Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Guiqiong He
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| |
Collapse
|
105
|
Carretero VJ, Álvarez-Merz I, Hernández-Campano J, Kirov SA, Hernández-Guijo JM. Targeting harmful effects of non-excitatory amino acids as an alternative therapeutic strategy to reduce ischemic damage. Neural Regen Res 2025; 20:2454-2463. [PMID: 39314160 PMCID: PMC11801293 DOI: 10.4103/nrr.nrr-d-24-00536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/16/2024] [Accepted: 08/14/2024] [Indexed: 09/25/2024] Open
Abstract
The involvement of the excitatory amino acids glutamate and aspartate in cerebral ischemia and excitotoxicity is well-documented. Nevertheless, the role of non-excitatory amino acids in brain damage following a stroke or brain trauma remains largely understudied. The release of amino acids by necrotic cells in the ischemic core may contribute to the expansion of the penumbra. Our findings indicated that the reversible loss of field excitatory postsynaptic potentials caused by transient hypoxia became irreversible when exposed to a mixture of just four non-excitatory amino acids (L-alanine, glycine, L-glutamine, and L-serine) at their plasma concentrations. These amino acids induce swelling in the somas of neurons and astrocytes during hypoxia, along with permanent dendritic damage mediated by N-methyl-D-aspartate receptors. Blocking N-methyl-D-aspartate receptors prevented neuronal damage in the presence of these amino acids during hypoxia. It is likely that astroglial swelling caused by the accumulation of these amino acids via the alanine-serine-cysteine transporter 2 exchanger and system N transporters activates volume-regulated anion channels, leading to the release of excitotoxins and subsequent neuronal damage through N-methyl-D-aspartate receptor activation. Thus, previously unrecognized mechanisms involving non-excitatory amino acids may contribute to the progression and expansion of brain injury in neurological emergencies such as stroke and traumatic brain injury. Understanding these pathways could highlight new therapeutic targets to mitigate brain injury.
Collapse
Affiliation(s)
| | - Iris Álvarez-Merz
- Department of Pharmacology and Therapeutic, School of Medicine, Univ. Autónoma de Madrid, Madrid, Spain
- Ramón y Cajal Institute for Health Research (IRYCIS), Neurobiology-Research Service, Hospital Ramón y Cajal, Madrid, Spain
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jorge Hernández-Campano
- Department of Pharmacology and Therapeutic, School of Medicine, Univ. Autónoma de Madrid, Madrid, Spain
| | - Sergei A. Kirov
- Department of Neuroscience and Regenerative Medicine & Department of Neurosurgery, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Jesús M. Hernández-Guijo
- Department of Pharmacology and Therapeutic, School of Medicine, Univ. Autónoma de Madrid, Madrid, Spain
- Ramón y Cajal Institute for Health Research (IRYCIS), Neurobiology-Research Service, Hospital Ramón y Cajal, Madrid, Spain
| |
Collapse
|
106
|
Li Z, Wu Y, Manyande A, Wu D, Xiang H. Odorgenetics with 2-pentanone: a novel cell manipulation technique. Med Gas Res 2025; 15:450-451. [PMID: 40072256 PMCID: PMC12054665 DOI: 10.4103/mgr.medgasres-d-25-00014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 01/27/2025] [Accepted: 02/15/2025] [Indexed: 04/20/2025] Open
Affiliation(s)
- Zhixiao Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Hubei Province, China
| | - Yanqiong Wu
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Institute of Anesthesiology & Pain (IAP), Department of Anesthesiology, Taihe Hospital, College of Pharmacy, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Anne Manyande
- School of Human and Social Sciences, University of West London, London, UK
| | - Duozhi Wu
- Department of Anesthesiology, Hainan General Hospital (Hainan Hospital Affiliated to Hainan Medical University), Haikou, Hainan Province, China
| | - Hongbing Xiang
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
107
|
Hashemolhosseini S, Gessler L. Crosstalk among canonical Wnt and Hippo pathway members in skeletal muscle and at the neuromuscular junction. Neural Regen Res 2025; 20:2464-2479. [PMID: 39248171 PMCID: PMC11801303 DOI: 10.4103/nrr.nrr-d-24-00417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/04/2024] [Accepted: 08/05/2024] [Indexed: 09/10/2024] Open
Abstract
Skeletal muscles are essential for locomotion, posture, and metabolic regulation. To understand physiological processes, exercise adaptation, and muscle-related disorders, it is critical to understand the molecular pathways that underlie skeletal muscle function. The process of muscle contraction, orchestrated by a complex interplay of molecular events, is at the core of skeletal muscle function. Muscle contraction is initiated by an action potential and neuromuscular transmission requiring a neuromuscular junction. Within muscle fibers, calcium ions play a critical role in mediating the interaction between actin and myosin filaments that generate force. Regulation of calcium release from the sarcoplasmic reticulum plays a key role in excitation-contraction coupling. The development and growth of skeletal muscle are regulated by a network of molecular pathways collectively known as myogenesis. Myogenic regulators coordinate the differentiation of myoblasts into mature muscle fibers. Signaling pathways regulate muscle protein synthesis and hypertrophy in response to mechanical stimuli and nutrient availability. Several muscle-related diseases, including congenital myasthenic disorders, sarcopenia, muscular dystrophies, and metabolic myopathies, are underpinned by dysregulated molecular pathways in skeletal muscle. Therapeutic interventions aimed at preserving muscle mass and function, enhancing regeneration, and improving metabolic health hold promise by targeting specific molecular pathways. Other molecular signaling pathways in skeletal muscle include the canonical Wnt signaling pathway, a critical regulator of myogenesis, muscle regeneration, and metabolic function, and the Hippo signaling pathway. In recent years, more details have been uncovered about the role of these two pathways during myogenesis and in developing and adult skeletal muscle fibers, and at the neuromuscular junction. In fact, research in the last few years now suggests that these two signaling pathways are interconnected and that they jointly control physiological and pathophysiological processes in muscle fibers. In this review, we will summarize and discuss the data on these two pathways, focusing on their concerted action next to their contribution to skeletal muscle biology. However, an in-depth discussion of the non-canonical Wnt pathway, the fibro/adipogenic precursors, or the mechanosensory aspects of these pathways is not the focus of this review.
Collapse
Affiliation(s)
- Said Hashemolhosseini
- Institute of Biochemistry, Medical Faculty, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Lea Gessler
- Institute of Biochemistry, Medical Faculty, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
108
|
Goncalves K, Przyborski S. Modulation of the Nogo signaling pathway to overcome amyloid-β-mediated neurite inhibition in human pluripotent stem cell-derived neurites. Neural Regen Res 2025; 20:2645-2654. [PMID: 39105379 PMCID: PMC11801276 DOI: 10.4103/nrr.nrr-d-23-01628] [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: 09/27/2023] [Revised: 05/29/2024] [Accepted: 07/09/2024] [Indexed: 08/07/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202509000-00026/figure1/v/2024-11-05T132919Z/r/image-tiff Neuronal cell death and the loss of connectivity are two of the primary pathological mechanisms underlying Alzheimer's disease. The accumulation of amyloid-β peptides, a key hallmark of Alzheimer's disease, is believed to induce neuritic abnormalities, including reduced growth, extension, and abnormal growth cone morphology, all of which contribute to decreased connectivity. However, the precise cellular and molecular mechanisms governing this response remain unknown. In this study, we used an innovative approach to demonstrate the effect of amyloid-β on neurite dynamics in both two-dimensional and three-dimensional culture systems, in order to provide more physiologically relevant culture geometry. We utilized various methodologies, including the addition of exogenous amyloid-β peptides to the culture medium, growth substrate coating, and the utilization of human-induced pluripotent stem cell technology, to investigate the effect of endogenous amyloid-β secretion on neurite outgrowth, thus paving the way for potential future applications in personalized medicine. Additionally, we also explore the involvement of the Nogo signaling cascade in amyloid-β-induced neurite inhibition. We demonstrate that inhibition of downstream ROCK and RhoA components of the Nogo signaling pathway, achieved through modulation with Y-27632 (a ROCK inhibitor) and Ibuprofen (a Rho A inhibitor), respectively, can restore and even enhance neuronal connectivity in the presence of amyloid-β. In summary, this study not only presents a novel culture approach that offers insights into the biological process of neurite growth and inhibition, but also proposes a specific mechanism for reduced neural connectivity in the presence of amyloid-β peptides, along with potential intervention points to restore neurite growth. Thereby, we aim to establish a culture system that has the potential to serve as an assay for measuring preclinical, predictive outcomes of drugs and their ability to promote neurite outgrowth, both generally and in a patient-specific manner.
Collapse
Affiliation(s)
| | - Stefan Przyborski
- Department of Biosciences, Durham University, Durham, UK
- Reprocell Europe Ltd., Glasgow, UK
| |
Collapse
|
109
|
Mukherjee TR, Tyagi O, Mehta RK. Differences in neural strategies explain exoskeleton-related benefits in performance over time during complex visuomotor wiring tasks. APPLIED ERGONOMICS 2025; 127:104532. [PMID: 40239298 DOI: 10.1016/j.apergo.2025.104532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 04/06/2025] [Accepted: 04/09/2025] [Indexed: 04/18/2025]
Abstract
This study evaluated the effects of a passive shoulder exoskeleton on neural, muscular, and perceptual responses during a three-day visuomotor wiring task involving overhead reaching. The task required participants to sequentially connect 40 randomly arranged targets on a two-dimensional board using a wire, engaging both motor and cognitive processes such as working memory and visual search. Twenty-four novice participants, balanced by sex, were randomly assigned to either an exoskeleton or a control group. The exoskeleton group demonstrated reduced upper extremity muscle activity but increased lower back activity. Despite similar perceived exertion levels between the groups, exoskeleton users reported lower mental demands, quicker visual searches, and higher task accuracy over time, all while exhibiting comparable task completion times. Neural analysis revealed greater functional specialization in the exoskeleton group, whereas the control group prioritized fronto-motor network integration. These findings provide valuable insights for practitioners contemplating the implementation of exoskeletons for tasks requiring both physical and cognitive engagement effort.
Collapse
Affiliation(s)
- Tiash Rana Mukherjee
- J. Mike Walker '66 Department of Mechanical Engineering, Texas A&M University, College Station, TX, 77840, USA
| | - Oshin Tyagi
- Department of Industrial & Operations Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ranjana K Mehta
- Department of Industrial Engineering, University of Wisconsin - Madison, Madison, WI, 53706, USA.
| |
Collapse
|
110
|
Wheeler DW, Ascoli GA. A novel method for clustering cellular data to improve classification. Neural Regen Res 2025; 20:2697-2705. [PMID: 39314166 PMCID: PMC11801281 DOI: 10.4103/nrr.nrr-d-24-00532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/15/2024] [Accepted: 08/15/2024] [Indexed: 09/25/2024] Open
Abstract
Many fields, such as neuroscience, are experiencing the vast proliferation of cellular data, underscoring the need for organizing and interpreting large datasets. A popular approach partitions data into manageable subsets via hierarchical clustering, but objective methods to determine the appropriate classification granularity are missing. We recently introduced a technique to systematically identify when to stop subdividing clusters based on the fundamental principle that cells must differ more between than within clusters. Here we present the corresponding protocol to classify cellular datasets by combining data-driven unsupervised hierarchical clustering with statistical testing. These general-purpose functions are applicable to any cellular dataset that can be organized as two-dimensional matrices of numerical values, including molecular, physiological, and anatomical datasets. We demonstrate the protocol using cellular data from the Janelia MouseLight project to characterize morphological aspects of neurons.
Collapse
Affiliation(s)
- Diek W. Wheeler
- Center for Neural Informatics, Structures, & Plasticity, Krasnow Institute for Advanced Study; and Bioengineering Department, Volgenau School of Engineering; George Mason University, Fairfax, VA, USA
| | - Giorgio A. Ascoli
- Center for Neural Informatics, Structures, & Plasticity, Krasnow Institute for Advanced Study; and Bioengineering Department, Volgenau School of Engineering; George Mason University, Fairfax, VA, USA
| |
Collapse
|
111
|
Alfahel L, Rajkovic A, Israelson A. Translational challenges in amyotrophic lateral sclerosis therapy with macrophage migration inhibitory factor. Neural Regen Res 2025; 20:2583-2584. [PMID: 39503423 PMCID: PMC11801292 DOI: 10.4103/nrr.nrr-d-24-00616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 02/08/2025] Open
Affiliation(s)
- Leenor Alfahel
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel; The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Aleksandar Rajkovic
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel; The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Adrian Israelson
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel; The School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
112
|
Franco R, Garrigós C, Capó T, Serrano-Marín J, Rivas-Santisteban R, Lillo J. Olfactory receptors in neural regeneration in the central nervous system. Neural Regen Res 2025; 20:2480-2494. [PMID: 39503417 PMCID: PMC11801295 DOI: 10.4103/nrr.nrr-d-24-00495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/05/2024] [Accepted: 08/05/2024] [Indexed: 02/08/2025] Open
Abstract
Olfactory receptors are crucial for detecting odors and play a vital role in our sense of smell, influencing behaviors from food choices to emotional memories. These receptors also contribute to our perception of flavor and have potential applications in medical diagnostics and environmental monitoring. The ability of the olfactory system to regenerate its sensory neurons provides a unique model to study neural regeneration, a phenomenon largely absent in the central nervous system. Insights gained from how olfactory neurons continuously replace themselves and reestablish functional connections can provide strategies to promote similar regenerative processes in the central nervous system, where damage often results in permanent deficits. Understanding the molecular and cellular mechanisms underpinning olfactory neuron regeneration could pave the way for developing therapeutic approaches to treat spinal cord injuries and neurodegenerative diseases like Alzheimer's disease. Olfactory receptors are found in almost any cell of every organ/tissue of the mammalian body. This ectopic expression provides insights into the chemical structures that can activate olfactory receptors. In addition to odors, olfactory receptors in ectopic expression may respond to endogenous compounds and molecules produced by mucosal colonizing microbiota. The analysis of the function of olfactory receptors in ectopic expression provides valuable information on the signaling pathway engaged upon receptor activation and the receptor's role in proliferation and cell differentiation mechanisms. This review explores the ectopic expression of olfactory receptors and the role they may play in neural regeneration within the central nervous system, with particular attention to compounds that can activate these receptors to initiate regenerative processes. Evidence suggests that olfactory receptors could serve as potential therapeutic targets for enhancing neural repair and recovery following central nervous system injuries.
Collapse
Affiliation(s)
- Rafael Franco
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- CiberNed Network Center for Biomedical Research in Neurodegenerative Diseases, Spanish National Health Institute Carlos III, Madrid, Spain
- School of Chemistry, Universitat de Barcelona, Barcelona, Spain
| | - Claudia Garrigós
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Toni Capó
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain
| | - Joan Serrano-Marín
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Rafael Rivas-Santisteban
- CiberNed Network Center for Biomedical Research in Neurodegenerative Diseases, Spanish National Health Institute Carlos III, Madrid, Spain
- Laboratory of Computational Medicine, Biostatistics Unit, Faculty of Medicine, Autonomous University of Barcelona, Campus Bellaterra, Barcelona, Spain
| | - Jaume Lillo
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- CiberNed Network Center for Biomedical Research in Neurodegenerative Diseases, Spanish National Health Institute Carlos III, Madrid, Spain
| |
Collapse
|
113
|
Pham TNM, Behl C. Cellular models of stress resistance may pave ways to fight neurodegenerative diseases. Neural Regen Res 2025; 20:2579-2580. [PMID: 39503421 PMCID: PMC11801301 DOI: 10.4103/nrr.nrr-d-24-00476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/01/2024] [Accepted: 08/15/2024] [Indexed: 02/08/2025] Open
Affiliation(s)
- Thu Nguyen Minh Pham
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Christian Behl
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
114
|
Lv Y, Li H. Blood diagnostic and prognostic biomarkers in amyotrophic lateral sclerosis. Neural Regen Res 2025; 20:2556-2570. [PMID: 39314138 PMCID: PMC11801290 DOI: 10.4103/nrr.nrr-d-24-00286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/23/2024] [Accepted: 09/06/2024] [Indexed: 09/25/2024] Open
Abstract
Amyotrophic lateral sclerosis is a devastating neurodegenerative disease for which the current treatment approaches remain severely limited. The principal pathological alterations of the disease include the selective degeneration of motor neurons in the brain, brainstem, and spinal cord, as well as abnormal protein deposition in the cytoplasm of neurons and glial cells. The biological markers under extensive scrutiny are predominantly located in the cerebrospinal fluid, blood, and even urine. Among these biomarkers, neurofilament proteins and glial fibrillary acidic protein most accurately reflect the pathologic changes in the central nervous system, while creatinine and creatine kinase mainly indicate pathological alterations in the peripheral nerves and muscles. Neurofilament light chain levels serve as an indicator of neuronal axonal injury that remain stable throughout disease progression and are a promising diagnostic and prognostic biomarker with high specificity and sensitivity. However, there are challenges in using neurofilament light chain to differentiate amyotrophic lateral sclerosis from other central nervous system diseases with axonal injury. Glial fibrillary acidic protein predominantly reflects the degree of neuronal demyelination and is linked to non-motor symptoms of amyotrophic lateral sclerosis such as cognitive impairment, oxygen saturation, and the glomerular filtration rate. TAR DNA-binding protein 43, a pathological protein associated with amyotrophic lateral sclerosis, is emerging as a promising biomarker, particularly with advancements in exosome-related research. Evidence is currently lacking for the value of creatinine and creatine kinase as diagnostic markers; however, they show potential in predicting disease prognosis. Despite the vigorous progress made in the identification of amyotrophic lateral sclerosis biomarkers in recent years, the quest for definitive diagnostic and prognostic biomarkers remains a formidable challenge. This review summarizes the latest research achievements concerning blood biomarkers in amyotrophic lateral sclerosis that can provide a more direct basis for the differential diagnosis and prognostic assessment of the disease beyond a reliance on clinical manifestations and electromyography findings.
Collapse
Affiliation(s)
- Yongting Lv
- Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Hongfu Li
- Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Department of Medical Genetics and Center for Rare disease, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Nanhu Brain-Computer Interface Institute, Hangzhou, Zhejiang Province, China
| |
Collapse
|
115
|
Chen TY, Xu J, Tai CH, Wen TK, Hsu SH. Biodegradable, electroconductive self-healing hydrogel based on polydopamine-coated polyurethane nano-crosslinker for Parkinson's disease therapy. Biomaterials 2025; 320:123268. [PMID: 40121830 DOI: 10.1016/j.biomaterials.2025.123268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/19/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by loss of dopaminergic neurons, causing motor and neurological impairments. Current treatments offer only temporary symptom relief without halting progression. Herein, a fully biodegradable, electroconductive self-healing hydrogel (CPUD gel) is developed, incorporating electroconductive polydopamine-coated polyurethane nanoparticles (PUD) as crosslinker. The core-shell PUD nanoparticles have a highly uniform size of ∼36 nm with a polydopamine shell of ∼4.8 nm thick on polyurethane core, revealed by small angle X-ray scattering, and own a conductivity of ∼0.82 mS/cm. As nano-crosslinker, the PUD can react with chitosan to form the dynamic CPUD hydrogel with shear modulus (∼280 Pa) and conductivity (∼4.34 mS/cm), mimicking brain tissue properties. In vitro, CPUD gel supports neural stem cell (NSC) proliferation (∼565 %) and differentiation, with elevated neuronal marker expression at 14 days, while exhibiting strong antioxidative and anti-inflammatory effects, rescuing ∼88 % inflamed NSCs. A therapeutic strategy combining injectable CPUD gel with acupuncture in a PD rat model, aiming to activate the innate regenerative mechanisms of body through mobilized endogenous stem cells, is further established. Using this approach, this hydrogel significantly elevates serum TGF-β1/SDF-1 levels, promotes dopaminergic neuron regeneration (>80 %), modulates neuroinflammation through M1-to M2-microglia transition (∼12.6-fold M2/M1 ratio), and improves motor function (from 8 % to 37 % forelimb contacts) in 14 days. Particularly, the electrophysiological spike rate is recovered from 66 to 19 spikes/s, close to the healthy rate 15 spikes/s. The synergistic immunomodulation and neuroprotection highlight the potential of CPUD gel as an advanced therapeutic tool for PD.
Collapse
Affiliation(s)
- Tsai-Yu Chen
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan, ROC
| | - Junpeng Xu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan, ROC
| | - Chun-Hwei Tai
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Tsung-Kai Wen
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien, Taiwan, ROC
| | - Shan-Hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan, ROC.
| |
Collapse
|
116
|
Honeycutt SC, Gilles-Thomas EA, Lichte DD, McSain SL, Mukherjee A, Loney GC. Behavioral economics of polysubstance use: The role of orexin-1 receptors in nicotine-induced augmentation of synthetic opioid consumption. Neuropharmacology 2025; 274:110467. [PMID: 40246272 DOI: 10.1016/j.neuropharm.2025.110467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/02/2025] [Accepted: 04/12/2025] [Indexed: 04/19/2025]
Abstract
Nicotine and opioid use disorders are highly comorbid in clinical populations. Ongoing nicotine administration facilitates opioid consumption in both rodents and humans. Moreover, preclinical studies support that former exposure to nicotine solely during adolescence augments opioid consumption in adulthood similarly to acute nicotine administration. This suggests that developmental nicotine exposure persistently alters the neural substrates underlying motivation in a manner that resembles the acute pharmacological actions of nicotine. The orexin system mediates motivation to consume opioids in large part through signaling at orexin-1 receptors (ORX1Rs). Both developmental nicotine exposure and acute nicotine administration profoundly alter functioning of the orexin system which may mediate the reinforcing enhancing properties of nicotine. Here, we used behavioral economic procedures to generate demand curves for consumption of the synthetic, short-acting, μ-opioid receptor agonist remifentanil (RMF) in adulthood following prior adolescent nicotine exposure (ANE) and again following reintroduction of acute nicotine administration (ANA). We found that ANE was sufficient to augment multiple indices of the motivational value of RMF in adulthood and these effects were further exacerbated by ANA given during RMF self-administration sessions. Additionally, we demonstrate that systemic antagonism of ORX1Rs with SB-334867 is more efficacious in limiting motivation for RMF in nicotine-exposed rats relative to controls and this differential efficacy was even greater in ANA conditions relative to former ANE. These findings support that nicotine-induced facilitation of orexin signaling may mechanistically contribute to augmented opioid consumption offering critical insight for treatment options for a population that is particularly vulnerable to developing opioid use disorder.
Collapse
Affiliation(s)
- Sarah C Honeycutt
- Department of Psychology, Program in Behavioral Neuroscience, The State University of New York University at Buffalo, Buffalo, NY, USA
| | - Elizabeth A Gilles-Thomas
- Department of Psychology, Program in Behavioral Neuroscience, The State University of New York University at Buffalo, Buffalo, NY, USA
| | - David D Lichte
- Department of Psychology, Program in Behavioral Neuroscience, The State University of New York University at Buffalo, Buffalo, NY, USA
| | - Shannon L McSain
- Department of Psychology, Program in Behavioral Neuroscience, The State University of New York University at Buffalo, Buffalo, NY, USA
| | - Ashmita Mukherjee
- Department of Psychology, Program in Behavioral Neuroscience, The State University of New York University at Buffalo, Buffalo, NY, USA
| | - Gregory C Loney
- Department of Psychology, Program in Behavioral Neuroscience, The State University of New York University at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
117
|
Sharpe AL, Liter LR, Donohue D, Carter KA, Vangeneugden P, Weaver SM, Stout MB, Beckstead MJ. Aged mice exhibit faster acquisition of intravenous opioid self-administration with variable effects on intake. Neuropharmacology 2025; 274:110464. [PMID: 40228626 DOI: 10.1016/j.neuropharm.2025.110464] [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: 09/24/2024] [Revised: 01/06/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
Although opioid abuse is more prevalent in young individuals, the rates of opioid use, overdose, and use disorders continue to climb among the elderly. Little is known about the biology underlying abuse potential in a healthy, aged population, in part due to technical and logistical difficulties testing intravenous self-administration in aged rodents. The goal of this study was to address a critical gap in the literature regarding age-dependent effects in opioid (remifentanil and fentanyl) self-administration. Male and female C57Bl/6J and C57Bl/6NJ mice were divided into young (mean: 19 weeks) and old (mean: 101 weeks) groups and were trained to self-administer intravenous fentanyl or remifentanil in daily operant sessions. Acquisition, intake, and cue-responding after forced abstinence were measured for both drugs, and a dose-response curve and dose-escalation were conducted for remifentanil and fentanyl, respectively. Surprisingly, old mice learned to self-administer both remifentanil and fentanyl faster and more accurately than young mice. Baseline intake of remifentanil was also greater in old mice compared to the young group; however, we did not see increased intake of fentanyl with age at either dose tested. Furthermore, old mice showed greater responding for cues previously associated with remifentanil after a forced abstinence, but this result was not observed with fentanyl. This first report of opioid self-administration in greater than 20-month-old mice suggests that old mice have an increased vulnerability for opioid use compared to younger counterparts, underscoring the importance of future work to uncover the biological mechanisms that are responsible.
Collapse
Affiliation(s)
- Amanda L Sharpe
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences, Oklahoma City, OK, USA.
| | - Laci R Liter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Graduate Program in Biomedical Sciences, University of Oklahoma Health Sciences, Oklahoma City, OK, USA
| | - Darius Donohue
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Kelsey A Carter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Patricia Vangeneugden
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Sofia M Weaver
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Michael B Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Michael J Beckstead
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
118
|
Wang W, Thomas ER, Xiao R, Chen T, Guo Q, Liu K, Yang Y, Li X. Targeting mitochondria-regulated ferroptosis: A new frontier in Parkinson's disease therapy. Neuropharmacology 2025; 274:110439. [PMID: 40174689 DOI: 10.1016/j.neuropharm.2025.110439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 03/16/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantial nigra. Mitochondrial dysfunction and mitochondrial oxidative stress are central to the pathogenesis of PD, with recent evidence highlighting the role of ferroptosis - a type of regulated cell death dependent on iron metabolism and lipid peroxidation. Mitochondria, the central organelles for cellular energy metabolism, play a pivotal role in PD pathogenesis through the production of Reactive oxygen species (ROS) and the disruption of iron homeostasis. This review explores the intricate interplay between mitochondrial dysfunction and ferroptosis in PD, focusing on key processes such as impaired electron transport chain function, tricarboxylic acid (TCA) cycle dysregulation, disruption of iron metabolism, and altered lipid peroxidation. We discuss key pathways, including the role of glutathione (GSH), mitochondrial ferritin, and the regulation of the mitochondrial labile iron pool (mLIP), which collectively influence the susceptibility of neurons to ferroptosis. Furthermore, this review emphasizes the importance of mitochondrial quality control mechanisms, such as mitophagy and mitochondrial biogenesis, in mitigating ferroptosis-induced neuronal death. Understanding these mechanisms linking the interplay between mitochondrial dysfunction and ferroptosis may pave the way for novel therapeutic approaches aimed at preserving mitochondrial integrity and preventing neuronal loss in PD.
Collapse
Affiliation(s)
- Wenjun Wang
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | | | - Ruyue Xiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Tianshun Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Qulian Guo
- Department of Pediatrics, Birth Defects and Childhood Hematological Oncology Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Kezhi Liu
- The Zigong Affiliated of Hospital of Southwest Medical University, Zigong mental health Center, Zigong Institute of Brain Science, Zigong, Sichuan Province, 643020, China
| | - You Yang
- Department of Pediatrics, Birth Defects and Childhood Hematological Oncology Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Xiang Li
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China; The Zigong Affiliated of Hospital of Southwest Medical University, Zigong mental health Center, Zigong Institute of Brain Science, Zigong, Sichuan Province, 643020, China; Health Science Center, Xi'an Jiaotong University, 710061, China.
| |
Collapse
|
119
|
Teixeira GP, Rocha L, Faria RX. The impact of membrane receptors on modulating empathic pain. Neuropharmacology 2025; 274:110471. [PMID: 40254122 DOI: 10.1016/j.neuropharm.2025.110471] [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: 11/13/2024] [Revised: 03/28/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
Humans can estimate each other's pain and provide adapted care to reduce it. Empathetic skills are crucial for caregivers involved in pain management; consequently, educational programs and theories have emphasized the positive role of empathy in reducing pain intensity. It is also widely assumed that if caregivers lack empathy, they will underestimate pain intensity in their patients, and this unempathetic attitude can negatively influence pain intensity perception. Empathy for pain is thought to activate the affective‒motivational components of the pain matrix, which includes the anterior insula, middle and anterior cingulate cortices and amygdala, as indicated by functional magnetic resonance imaging and other methodologies. Activity in this core neural network reflects the affective experience that activates our responses to pain and lays the neural foundation for our understanding of our own emotions and those of others. Additionally, a variety of factors can regulate the intensity of empathy for pain, such as oxytocin and vasopressin receptors. Therefore, we selectively review the molecular mechanisms by which membrane receptors modulate this pain modality.
Collapse
Affiliation(s)
- Guilherme Pegas Teixeira
- Laboratory for Evaluation and Promotion of Environmental Health, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Post-Graduation Program in Vegetal Biotechnology and Bioprocesses, Rio de Janeiro Federal University, Rio de Janeiro, CEP, 21941-902, Brazil.
| | - Leandro Rocha
- Laboratory of Natural Products Technology, Faculty of Pharmacy, Fluminense Federal University, Rua Doutor Mário Viana 523, Santa Rosa, Niterói, CEP, 24241-002, Brazil; Post-Graduation Program in Vegetal Biotechnology and Bioprocesses, Rio de Janeiro Federal University, Rio de Janeiro, CEP, 21941-902, Brazil.
| | - Robson Xavier Faria
- Laboratory for Evaluation and Promotion of Environmental Health, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.
| |
Collapse
|
120
|
Guo L, Li X, Zhang R, Hou Y, Ma B, Li Z, Lv J, Wang B, Ma S, Li L, Yan L, Zhang B, Liu W, Lim K, Diao H, Wang S, Zhang C. In situ dual-activated NIRF/PA carrier-free nanoprobe for diagnosis and treatment of Parkinson's disease. Biosens Bioelectron 2025; 282:117473. [PMID: 40267542 DOI: 10.1016/j.bios.2025.117473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/09/2025] [Accepted: 04/12/2025] [Indexed: 04/25/2025]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease threatening the life of millions people worldwide. Oxidative stress, mitochondrial dysfunction, and neuroinflammation are the pivotal causative elements of PD. Precise diagnosis enables timely monitoring initiation and progression of PD, thereby facilitating the formulation of customized and targeted treatment strategies. Optical imaging offers one alternative way for PD diagnosis. However, available diagnostic probes suffer from the inability to bypass the blood brain barrier (BBB). To accurately diagnose and effectively combat PD, there is an urgent need to develop an integrated diagnostic and therapeutic nanoprobe that can bypass the BBB and target the factors underlying degeneration of dopaminergic (DA) neurons. In present study, one integrated carrier-free nanoprobe HVCur-NPs towards those factors was designed and constructed. By modifying probe side chain with polypeptide, RVG29, we obtained brain-targeting HV-PEG-RVG29. It not only enables BBB penetration, but also produces near-infrared fluorescence (NIRF) and photoacoustic (PA) signals in cascade response to H2O2 and viscosity. The release of loaded curcumin (CUR) prevents oxidative stress, neuroinflammation and restore mitochondrial function so as to rescue PD phenotypes. In cellular PD model, HVCur-NPs generated NIRF/PA signals in response to elevated ROS and viscosity, and ameliorated cell apoptosis by eliminating ROS and restoring mitochondria function. Moreover, in mice PD model, HVCur-NPs realized in situ NIRF/PA imaging brain, and rescued DA neuron loss and restored the behavioral deficit of PD mice, without detectable biotoxicity. This carrier-free nanoprobe opens venues for integrated diagnosis and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Lixia Guo
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, PR China; School of Pharmacy, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Xiaowan Li
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Run Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Yixuan Hou
- School of Pharmacy, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Bolong Ma
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Zheng Li
- Yong Loo Lin School of Medicine, National University of Singapore, 117054, Singapore
| | - Jiye Lv
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Bin Wang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Sufang Ma
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Lihong Li
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Lili Yan
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Boye Zhang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Wen Liu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, PR China
| | - Kahleong Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, 308232, Singapore
| | - Haipeng Diao
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, PR China.
| | - Shaowei Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan, 030001, PR China; The Second Clinical Medical College, Shanxi Medical University, Taiyuan, 030000, PR China.
| | - Chengwu Zhang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, PR China.
| |
Collapse
|
121
|
Contarino A. Genetic inactivation of the CRF 2 receptor eliminates morphine-induced sociability deficits in female mice. Neuropharmacology 2025; 274:110480. [PMID: 40274013 DOI: 10.1016/j.neuropharm.2025.110480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/14/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Social behavior deficits, such as poor sociability and social isolation, are key clinical features of substance use disorders. The corticotropin-releasing factor (CRF) system may underlie the effects of substances of abuse but its implication in substance-induced social behavior deficits remains largely unknown. CRF signaling is mediated by two receptor types, termed CRF1 and CRF2. Using the genetic mouse model of CRF2 receptor-deficiency and the three-chamber task for sociability, the present studies examined the specific role for the CRF2 receptor in sociability deficits induced by morphine. Notably, to assess possible sex-linked differences, female and male CRF2 receptor wild-type (CRF2 WT) or knockout (CRF2 KO) mice were used. Intraperitoneal administration of morphine (1 mg/kg) reliably eliminated the preference for an unfamiliar same-sex conspecific over an object in female CRF2 WT, but not in CRF2 KO, mice, revealing a key role for the CRF2 receptor in opiate-induced sociability deficits. In contrast, morphine almost significantly and similarly reduced the preference for an unfamiliar same-sex conspecific over an object in male CRF2 WT and CRF2 KO mice, indicating no role for the CRF2 receptor. Notably, treatment with morphine never affected distance travelled during the three-chamber test, indicating that CRF2 receptor-dependent or -independent opiate effects were specific to social behavior. The present findings provide initial evidence of a critical sex-linked role for the CRF2 receptor in social behavior deficits induced by opiate substances, suggesting new, sex-customized, therapeutic strategy for opioid use disorders.
Collapse
Affiliation(s)
- Angelo Contarino
- Université Paris Cité, INSERM, CNRS, Health & Functional Exposomics - HealthFex, U1124, 75006, Paris, France; Université de Bordeaux, 33076, Bordeaux, France.
| |
Collapse
|
122
|
Wang Q, Bie Y, Xia X, Liu Y, Blank I, Shi Y, Men H, Chen YP. Mechanistic study of saltiness enhancement induced by three characteristic volatiles identified in Jinhua dry-cured ham using electroencephalography (EEG). Food Chem 2025; 482:144180. [PMID: 40199153 DOI: 10.1016/j.foodchem.2025.144180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/05/2025] [Accepted: 03/31/2025] [Indexed: 04/10/2025]
Abstract
Excessive salt intake is a pressing food health issue, and odor-induced saltiness enhancement (OISE) is a novel strategy for targeted salt reduction. Understanding the neural mechanisms of OISE is essential for salt reduction. In this study, the mechanism of saltiness enhancement induced by three volatile organic compounds (VOCs) identified in Jinhua dry ham was investigated in 20 panelists using electroencephalography (EEG). The study demonstrated that VOCs enhanced salty taste perception, primarily through low-frequency brain waves. Source localization revealed occipital lobe activation during salty taste recognition, while OISE stimuli enhanced activity in the primary and secondary gustatory cortices. Additionally, VOCs enhanced phase synchronization among activated brain regions, as indicated by functional connectivity. This study enhances the understanding of olfactory-gustatory interactions and provides a neurological basis for the effects of OISE.
Collapse
Affiliation(s)
- Qun Wang
- School of Automation Engineering, Northeast Electric Power University, Jilin 132012, China.
| | - Yongjing Bie
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Xiuxin Xia
- School of Automation Engineering, Northeast Electric Power University, Jilin 132012, China.
| | - Yuan Liu
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China; School of Food Science and Engineering, Ningxia University, Yinchuan 750021, China.
| | - Imre Blank
- School of Food Science and Engineering, Ningxia University, Yinchuan 750021, China.
| | - Yan Shi
- School of Automation Engineering, Northeast Electric Power University, Jilin 132012, China.
| | - Hong Men
- School of Automation Engineering, Northeast Electric Power University, Jilin 132012, China.
| | - Yan Ping Chen
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
123
|
de Hevia MD, Veggiotti L, Baqqali Y. Spatial associations of number and pitch in human newborns. J Exp Child Psychol 2025; 256:106259. [PMID: 40273466 DOI: 10.1016/j.jecp.2025.106259] [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: 10/09/2024] [Revised: 03/21/2025] [Accepted: 03/21/2025] [Indexed: 04/26/2025]
Abstract
Humans use space to think, reason about, externally represent, and even talk about many dimensions (e.g., time, pitch height). One dimension that appears to exploit spatial resources is the mental representation of the numerosity of a set in the form of a mental number line. Although the horizontal number-space mapping is present from birth (small-left vs. large-right), it is unknown whether it extends to other spatial axes from birth or whether it is later acquired through development/experience. Moreover, newborns map changes in pitch height onto a vertical axis (low pitch-bottom vs. high pitch-top), but it is an open question whether it extends to other spatial axes. We presented newborns (N = 64 total, n = 16 per experiment, 0-4 days) with an auditory increase/decrease in magnitude along with a visual figure on a vertically oriented screen (bottom vs. top, change in number: Experiments 1 and 2; change in pitch: Experiment 3) or on a horizontally oriented screen (left vs. right, change in pitch: Experiment 4). Newborns associated changes in magnitude with a vertical axis only when experiencing an increase in magnitude (increase/up); however, the possibility that visuospatial biases could account for this asymmetric pattern are discussed. Newborns did not map changes in pitch horizontally (Experiment 4), in line with previous work showing that the horizontal mapping of number at birth does not generalize to other dimensions. These findings suggest that the flexible use of different spatial axes to map magnitude is not functional at birth and that the horizontal mapping of number might be privileged.
Collapse
Affiliation(s)
- Maria Dolores de Hevia
- Université Paris Cité, CNRS, Integrative Neuroscience and Cognition Center, 75006 Paris, France.
| | - Ludovica Veggiotti
- Université Paris Cité, CNRS, Integrative Neuroscience and Cognition Center, 75006 Paris, France; Université Paris Cité, Institut Pasteur, AP-HP, Inserm, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, 75012 Paris, France
| | - Yasmine Baqqali
- Université Paris Cité, CNRS, Integrative Neuroscience and Cognition Center, 75006 Paris, France
| |
Collapse
|
124
|
Hayat M, Syed RA, Qaiser H, Uzair M, Al-Regaiey K, Khallaf R, Albassam LAM, Kaleem I, Wang X, Wang R, Bhatti MS, Bashir S. Decoding molecular mechanisms: brain aging and Alzheimer's disease. Neural Regen Res 2025; 20:2279-2299. [PMID: 39104174 PMCID: PMC11759015 DOI: 10.4103/nrr.nrr-d-23-01403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 01/23/2024] [Accepted: 07/04/2024] [Indexed: 08/07/2024] Open
Abstract
The complex morphological, anatomical, physiological, and chemical mechanisms within the aging brain have been the hot topic of research for centuries. The aging process alters the brain structure that affects functions and cognitions, but the worsening of such processes contributes to the pathogenesis of neurodegenerative disorders, such as Alzheimer's disease. Beyond these observable, mild morphological shifts, significant functional modifications in neurotransmission and neuronal activity critically influence the aging brain. Understanding these changes is important for maintaining cognitive health, especially given the increasing prevalence of age-related conditions that affect cognition. This review aims to explore the age-induced changes in brain plasticity and molecular processes, differentiating normal aging from the pathogenesis of Alzheimer's disease, thereby providing insights into predicting the risk of dementia, particularly Alzheimer's disease.
Collapse
Affiliation(s)
- Mahnoor Hayat
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Rafay Ali Syed
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Hammad Qaiser
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad (IIUI), Islamabad, Pakistan
| | - Mohammad Uzair
- Department of Bioengineering, King Fahd University of Petroleum and Minerals, Dhahran, Saudi Arabia
| | - Khalid Al-Regaiey
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Roaa Khallaf
- Department of Neurology, Neuroscience Center, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | | | - Imdad Kaleem
- Department of Biosciences, Commission on Science and Technology for Sustainable Development in the South (COMSATS University), Islamabad, Pakistan
| | - Xueyi Wang
- Department of Psychiatry, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
- Mental Health Institute of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Ran Wang
- Department of Psychiatry, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
- Mental Health Institute of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Mehwish S. Bhatti
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Shahid Bashir
- Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam, Saudi Arabia
| |
Collapse
|
125
|
Yoon D, Lee H. In silico discovery of novel compounds for FAK activation using virtual screening, AI-based prediction, and molecular dynamics. Comput Biol Chem 2025; 117:108420. [PMID: 40157227 DOI: 10.1016/j.compbiolchem.2025.108420] [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: 11/24/2024] [Revised: 02/19/2025] [Accepted: 02/28/2025] [Indexed: 04/01/2025]
Abstract
Focal Adhesion Kinase (FAK) is a non-receptor tyrosine kinase that plays a crucial role in cell proliferation, migration, and signal transduction. FAK is overexpressed in metastatic and advanced-stage cancers, where it is considered a key kinase in cancer growth and metastasis. However, recent research has revealed that FAK activity decreases in various diseases. we aimed to identify compounds that could enhance FAK activity using structure-based virtual screening and artificial intelligence models from a vast chemical database. We began with an extensive chemical database containing over 10 million compounds and used our newly developed pipeline to screen candidate molecules. To select compounds structurally similar to ZINC40099027 (ZN27), a known FAK activator, we calculated Tanimoto Similarity scores and chose compounds with a score of at least 0.8. Clustering was performed using K-means based on the molecular properties. Subsequently, we utilized docking simulation, deep learning and SAScorer to evaluate and predict the protein-ligand docking affinity and physicochemical properties of the candidate compounds. The deep learning models were selected as state-of-the-art models: GLAM predicts the blood-brain barrier permeability of FAK, and elEmBERT predicts the potential toxicity of compound. The combined results were used to create an evaluation matrix. We selected 10 promising candidate compounds from the initial dataset of 10 million. To evaluate the stability of these top 10 candidate compounds in interaction with the FAK protein, we conducted Molecular Dynamics (MD) simulations. We performed a molecular dynamics simulation for a total of 50 ns and identified the top three promising candidate compounds.
Collapse
Affiliation(s)
- Deokhyeon Yoon
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Hyunsu Lee
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea; Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, 50612, Republic of Korea.
| |
Collapse
|
126
|
Liu X, Sanchez SW, Gong Y, Riddle R, Jiang Z, Trevor S, Contag CH, Saha D, Li W. An insect-based bioelectronic sensing system combining flexible dual-sided microelectrode array and insect olfactory circuitry for human lung cancer detection. Biosens Bioelectron 2025; 281:117356. [PMID: 40215892 DOI: 10.1016/j.bios.2025.117356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 05/04/2025]
Abstract
Early detection of lung cancer significantly enhances treatment outcomes, yet current screening methods are limited by accessibility, sensitivity, and cost. This study introduces a bioelectronic sensing platform that integrates the highly sensitive locust olfactory system with a flexible dual-sided microelectrode array (MEA), for robust, noninvasive, and label-free detection of volatile lung cancer biomarkers. Using an innovative folding-annealing fabrication technique and PEDOT:PSS surface functionalization, we developed flexible, dual-sided MEAs with high electrode densities of 463, 687, and 766 channels/mm2 across prototypes, maintaining low impedance (within 4 × 104 Ω). These MEAs demonstrated mechanical flexibility and stability, enabling direct insertion into locust brain tissue without mechanical reinforcement and facilitating precise recording of neural activity in the antennal lobe triggered by lung cancer-related volatile organic compounds (VOCs) from low concentration (1 ppm). Advanced dimensionality reduction techniques applied to the electrophysiological recordings identified distinct neural response patterns to each VOC biomarker and the complex "scent" emitted from various cell lines. Using high-dimensional population neuronal response analysis with a leave-one-trial-out approach, the platform achieved a 100 % classification success rate for unknown VOCs. Additionally, varying concentrations (ppm-ppb) of individual VOC biomarkers were detected and classified with an accuracy of 86 %. The system was further tested for its ability to detect and classify human lung cancer cell lines based on the unique "scent" of cultured cells, including two non-small cell lung cancer (NSCLC) and two small cell lung cancer (SCLC) types. Quantitative assessments demonstrated that the platform achieved a classification accuracy of 85 % across these cell lines. These results substantiate the platform's potential for enhancing clinical diagnostics through the accurate identification of lung cancer stages and cell types. By integrating biological sensory systems with advanced bioelectronics, this study introduces a novel and efficient approach to lung cancer biomarker detection. It provides a non-invasive, brain-based cancer screening method, offering an accessible and innovative solution for early lung cancer diagnosis.
Collapse
Affiliation(s)
- Xiang Liu
- Neuroscience Program, Department of Physiology, Michigan State University, East Lansing, MI, USA; Institute for Quantitative Health Science and Engineering (IQ), East Lansing, MI, USA
| | - Simon W Sanchez
- Institute for Quantitative Health Science and Engineering (IQ), East Lansing, MI, USA; Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
| | - Yan Gong
- Institute for Quantitative Health Science and Engineering (IQ), East Lansing, MI, USA; Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI, USA
| | - Roksana Riddle
- Institute for Quantitative Health Science and Engineering (IQ), East Lansing, MI, USA; Department of Microbiology, Genetics & Immunology, Michigan State University, East Lansing, MI, USA
| | - Zebin Jiang
- Institute for Quantitative Health Science and Engineering (IQ), East Lansing, MI, USA; Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI, USA
| | - Stevens Trevor
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI, USA
| | - Christopher H Contag
- Institute for Quantitative Health Science and Engineering (IQ), East Lansing, MI, USA; Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA; Department of Microbiology, Genetics & Immunology, Michigan State University, East Lansing, MI, USA
| | - Debajit Saha
- Neuroscience Program, Department of Physiology, Michigan State University, East Lansing, MI, USA; Institute for Quantitative Health Science and Engineering (IQ), East Lansing, MI, USA; Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA.
| | - Wen Li
- Neuroscience Program, Department of Physiology, Michigan State University, East Lansing, MI, USA; Institute for Quantitative Health Science and Engineering (IQ), East Lansing, MI, USA; Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA; Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
127
|
Irie T, Matsuda T. In vivo direct neuronal conversion as a therapeutic strategy for ischemic stroke. Neural Regen Res 2025; 20:2309-2310. [PMID: 39359083 PMCID: PMC11759033 DOI: 10.4103/nrr.nrr-d-24-00545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/11/2024] [Accepted: 06/26/2024] [Indexed: 10/04/2024] Open
Affiliation(s)
- Takashi Irie
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Taito Matsuda
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
128
|
You X, Niu L, Fu J, Ge S, Shi J, Zhang Y, Zhuang P. Bidirectional regulation of the brain-gut-microbiota axis following traumatic brain injury. Neural Regen Res 2025; 20:2153-2168. [PMID: 39359076 PMCID: PMC11759007 DOI: 10.4103/nrr.nrr-d-24-00088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/20/2024] [Accepted: 05/11/2024] [Indexed: 10/04/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202508000-00002/figure1/v/2024-09-30T120553Z/r/image-tiff Traumatic brain injury is a prevalent disorder of the central nervous system. In addition to primary brain parenchymal damage, the enduring biological consequences of traumatic brain injury pose long-term risks for patients with traumatic brain injury; however, the underlying pathogenesis remains unclear, and effective intervention methods are lacking. Intestinal dysfunction is a significant consequence of traumatic brain injury. Being the most densely innervated peripheral tissue in the body, the gut possesses multiple pathways for the establishment of a bidirectional "brain-gut axis" with the central nervous system. The gut harbors a vast microbial community, and alterations of the gut niche contribute to the progression of traumatic brain injury and its unfavorable prognosis through neuronal, hormonal, and immune pathways. A comprehensive understanding of microbiota-mediated peripheral neuroimmunomodulation mechanisms is needed to enhance treatment strategies for traumatic brain injury and its associated complications. We comprehensively reviewed alterations in the gut microecological environment following traumatic brain injury, with a specific focus on the complex biological processes of peripheral nerves, immunity, and microbes triggered by traumatic brain injury, encompassing autonomic dysfunction, neuroendocrine disturbances, peripheral immunosuppression, increased intestinal barrier permeability, compromised responses of sensory nerves to microorganisms, and potential effector nuclei in the central nervous system influenced by gut microbiota. Additionally, we reviewed the mechanisms underlying secondary biological injury and the dynamic pathological responses that occur following injury to enhance our current understanding of how peripheral pathways impact the outcome of patients with traumatic brain injury. This review aimed to propose a conceptual model for future risk assessment of central nervous system-related diseases while elucidating novel insights into the bidirectional effects of the "brain-gut-microbiota axis."
Collapse
Affiliation(s)
- Xinyu You
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Niu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiafeng Fu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shining Ge
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiangwei Shi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yanjun Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Pengwei Zhuang
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
129
|
Qi S, Ritchie JL, Soto DA, Pruitt AY, Reeves DA, Artimenia LM, Fuchs RA. Sex-dependent role of the dorsolateral septum in shaping contextual cocaine memory strength. Neuropharmacology 2025; 273:110459. [PMID: 40204057 DOI: 10.1016/j.neuropharm.2025.110459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/05/2025] [Accepted: 04/06/2025] [Indexed: 04/11/2025]
Abstract
Established memories can be destabilized, updated, and reconsolidated into long-term memory stores. Memory updating and reconsolidation can alter the strength of maladaptive contextual drug memories and consequently context-induced drug craving and relapse. The dorsolateral septum (dlS) is a GABAergic nucleus that receives dense direct input from the cornu ammonis 3 regions of the dorsal hippocampus, a brain region that is critical for the maintenance of contextual cocaine memories. Accordingly, we tested the hypothesis that neuronal activity in the dlS regulates the strength of cocaine-predictive contextual memories prior to reconsolidation. Male and female Sprague-Dawley rats received cocaine self-administration training followed by extinction training in two different environmental contexts. After the last extinction training session, the rats were placed back into the cocaine-predictive context to retrieve and destabilize their cocaine-related contextual memories. Immediately or 6 h after memory retrieval, the rats received intra-dlS vehicle or baclofen/muscimol (B/M; GABAB/A agonists) infusions to inhibit neuronal activity during or after memory updating/reconsolidation, respectively. Resulting changes in cocaine and extinction memory strength were assessed based on the magnitude of unreinforced lever responding in the two contexts. Intra-dlS B/M infusion immediately after memory retrieval increased subsequent context-induced cocaine seeking behaviors in male rats, but not in female rats, whereas delayed B/M treatment had no effects in male rats. Together these findings suggest that the dlS is selectively engaged during memory updating/reconsolidation to reduce the strength of cocaine memories in males, possibly contributing to sex differences in the progression of cocaine use disorder.
Collapse
Affiliation(s)
- S Qi
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA
| | - J L Ritchie
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA
| | - D A Soto
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA
| | - A Y Pruitt
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA
| | - D A Reeves
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA
| | - L M Artimenia
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA
| | - R A Fuchs
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA; Washington State University Alcohol and Drug Abuse Research Program, Pullman, WA, USA.
| |
Collapse
|
130
|
Kim SH, Kim YH, Kim JB, Park NY, So JH, Park D, Choi DK, Yeom E, Gwon Y, Jo DS, Lee JA, Bae JE, Cho DH. Suppression of stress granule assembly by pyridoxal hydrochloride attenuates oxidative damage in skin fibroblasts. Bioorg Med Chem Lett 2025; 123:130238. [PMID: 40216204 DOI: 10.1016/j.bmcl.2025.130238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/28/2025] [Accepted: 04/03/2025] [Indexed: 04/17/2025]
Abstract
Stress granules (SGs) are membrane-less cytoplasmic structures that form in response to various stress stimuli and play a critical role in maintaining cellular homeostasis. Dysregulation of SG dynamics has been implicated in several diseases, including neurodegenerative and inflammatory conditions; however, their role in skin biology remains largely unexplored. In this study, we identified pyridoxal hydrochloride, a form of vitamin B6, as a novel regulator of SG formation through a metabolite library screening. Our results demonstrate that pyridoxal hydrochloride significantly suppresses oxidative stress-induced SG formation in skin fibroblasts, exhibiting effects comparable to G3Ia, a known SG inhibitor. Furthermore, pyridoxal hydrochloride mitigates oxidative stress by reducing reactive oxygen species (ROS) accumulation and preventing cell toxicity. Notably, it also attenuates ROS-induced upregulation of MMP1, thereby preserving collagen1 stability. These findings suggest the crucial role of SGs in skin fibroblast homeostasis and suggest that pyridoxal hydrochloride may serve as a potential therapeutic agent for oxidative stress-related skin disorders.
Collapse
Affiliation(s)
- Seong Hyun Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Yong Hwan Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Joon Bum Kim
- Organelle Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Na Yeon Park
- Organelle Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Jun Hee So
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Daeun Park
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Dong Kyu Choi
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Eunbyul Yeom
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Youngdae Gwon
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Doo Sin Jo
- Orgasis Corp., Suwon, Gyeonggi-do, Republic of Korea
| | - Jin-A Lee
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon, Republic of Korea
| | - Ji-Eun Bae
- Organelle Institute, Kyungpook National University, Daegu, Republic of Korea.
| | - Dong-Hyung Cho
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea; Organelle Institute, Kyungpook National University, Daegu, Republic of Korea; Orgasis Corp., Suwon, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
131
|
Petrescu TC, Błachnio A, Enea V. Internet gaming disorder in children and adolescents: A systematic review of familial protective and risk factors. Addict Behav 2025; 167:108345. [PMID: 40154103 DOI: 10.1016/j.addbeh.2025.108345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/27/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
Empirical research investigated psycho-social factors associated with the development and maintenance of Internet Gaming Disorder (IGD) in children and adolescents, but their potential role has not been highlighted in systematic reviews. The aims of the current systematic review were to (1) summarize and synthesize findings from empirical research on family factors related to children and adolescents' IGD; (2) identify familial protective and risk factors that are related to the development and maintenance of IGD in children and adolescents, and (3) provide suggestions for future research. A number of 64 studies fulfilled the inclusion criteria in the review from the following databases: Web of Science, Science Direct, Scopus, PubMed, ProQuest, Google Academic, and APA PsycNet. Family variables have been significantly connected to gaming addiction levels in children and adolescents. Results were divided into five main themes: parental mediation, positive parenting, poor parenting, familial disharmony, and familial socioeconomic status. Protective factors included parental knowledge and positive parenting, while poor parenting, familial disharmony and familial socioeconomic status with all their sub-themes represented risk factors. Restrictive mediation and affected parenting provided inconclusive results that deserve further research. Familial connection is imperative for gaming addiction prevention. A stressful familial environment (e.g., parental conflicts) could increase gaming addiction coping behavior. Longitudinal and cross-sectional results provided inconsistent results regarding the role of parental depression and parental mediation in children's gaming addiction development. Cross-cultural studies are needed on familial factors related to children's gaming addiction. Further longitudinal studies could provide answers for conflicting or underexplored areas.
Collapse
|
132
|
Kim M, López-Cano M, Zhang K, Wang Y, Gómez-Santacana X, Flores Á, Wu M, Li S, Zhang H, Wei Y, Li X, Good CH, Banks AR, Llebaria A, Hernando J, Sunwoo SH, Gu J, Huang Y, Ciruela F, Rogers JA. Wireless, battery-free, remote photoactivation of caged-morphine for photopharmacological pain modulation without side effects. Biosens Bioelectron 2025; 281:117440. [PMID: 40220492 DOI: 10.1016/j.bios.2025.117440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/26/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025]
Abstract
Chronic pain severely impairs physical, psychological, and cognitive functions. While opioid-based therapies can be effective, they are limited by tolerance, dependence, and adverse side effects, highlighting the need for safer alternatives. Recent advances in photopharmacology allow precise modulation of pain-related neuronal circuits, offering improved control and effectiveness. For delivery of light, fully implantable, wireless, battery-free optical systems in miniaturized forms offer attractive options relative to alternatives that use conventional bulk hardware and fiber optic tethers. This work presents a technology of this type, based on microscale light-emitting diodes (μ-ILEDs) and near-field communication (NFC) protocols, and optimized to activate photocaged morphine (pc-Mor) in targeted regions of the spinal cord. The unique flexible, lightweight designs ensure stable, minimally invasive operation in small animal model behavioral studies, with efficient power consumption and minimized thermal load on fragile tissues. Experimental results demonstrate effective pain suppression and reduced opioid-related side effects in an animal model of pain, thereby establishing this platform as a promising solution for chronic pain management.
Collapse
Affiliation(s)
- Minsung Kim
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, 60208, USA; Neurolux Inc., Northfield, IL, 60093, USA
| | - Marc López-Cano
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, 08907, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, 08907, Spain
| | - Kaiqing Zhang
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, 60208, USA; State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, Liaoning, 116024, China
| | - Yue Wang
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, 60208, USA; Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Xavier Gómez-Santacana
- MCS - Medicinal Chemistry & Synthesis, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - África Flores
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, 08907, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, 08907, Spain
| | - Mingzheng Wu
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, 60208, USA; Department of Neurobiology, Northwestern University, Evanston, IL, 60208, USA
| | - Shupeng Li
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Haohui Zhang
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Yuanting Wei
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Xiuyuan Li
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, 60208, USA; State Key Laboratory of Mechanical System and Vibration, School of Mechanical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Cameron H Good
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, 60208, USA; Neurolux Inc., Northfield, IL, 60093, USA; Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, 60208, USA
| | - Anthony R Banks
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, 60208, USA; Neurolux Inc., Northfield, IL, 60093, USA; Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, 60208, USA
| | - Amadeu Llebaria
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Jordi Hernando
- Departament de Química, Universitat Autònoma de Barcelona, Edifici C/n, Campus UAB, Cerdanyola del Vallès, 08193, Spain
| | - Sung-Hyuk Sunwoo
- Department of Chemical Engineering, Kumoh National Institute of Technology, Gumi, 39177, Republic of Korea
| | - Jianyu Gu
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, 60208, USA
| | - Yonggang Huang
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, 60208, USA; Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, 60208, USA; Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA; Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA.
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, 08907, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, 08907, Spain.
| | - John A Rogers
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, 60208, USA; Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA; Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, 60208, USA; Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA; Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA; Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
133
|
Guo X, Zheng Q, Gao W, Xiao Y, Shi L, Lin F, Xiong Y, Zhang Y, Xu Q, Wang L, He S, Zheng F. Synergistic microglial modulation by laminarin-based platinum nanozymes for potential intracerebral hemorrhage therapy. Biomaterials 2025; 319:123212. [PMID: 40031123 DOI: 10.1016/j.biomaterials.2025.123212] [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: 10/09/2024] [Revised: 01/26/2025] [Accepted: 02/23/2025] [Indexed: 03/05/2025]
Abstract
Abnormal microglial activation increases inflammation, causing significant brain damage after intracerebral hemorrhage (ICH). To aid recovery, treatments should regulate oxidative stress and inhibit the M1-like phenotype (pro-inflammation) of microglia. Recently, antioxidant nanozymes have emerged as tools for modulating microglial states, but detailed studies on their role in ICH treatment are limited. To address this, we developed an ultra-small (3-4 nm) laminarin-modified platinum nanozyme (Pt@LA) for the synergistic regulation of microglial polarization, offering a novel therapeutic strategy for ICH. Pt@LA effectively scavenges reactive oxygen species (ROS) through superoxide dismutase (SOD) and catalase (CAT)-like activities. Laminarin may inhibit the Dectin-1 receptor on microglia and its inflammatory pathway, Syk/NF-κB, reducing neuroinflammation. In vitro, Pt@LA decreased pro-inflammatory microglia and cytokine expression by inhibiting the Dectin-1/Syk/NF-κB and ROS-mediated NF-κB pathways. Furthermore, Pt@LA protected neurons, inhibited glial scar formation, and improved neurological function in ICH rats. Overall, this study presents Pt nanozymes based on naturally extracted laminarin and explores their application in alleviating oxidative stress and neuroinflammation after ICH, bridging nanozyme research and neuroscience.
Collapse
Affiliation(s)
- Xiumei Guo
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China; Laboratory of Clinical Pharmacy, Department of Pharmacy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China; Medical Center for Neurological Disorders, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China; Department of Neurology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Qionghua Zheng
- Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Department of Pharmaceutical Analysis, Fujian Medical University, Fuzhou, 350004, China
| | - Wen Gao
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China; Medical Center for Neurological Disorders, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China; Department of Neurology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Yinfeng Xiao
- Laboratory of Clinical Pharmacy, Department of Pharmacy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China; Medical Center for Neurological Disorders, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
| | - Liyong Shi
- Department of Pulmonary and Critical Care Medicine, Fujian Key Laboratory of Lung Stem Cell, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Fenglin Lin
- Laboratory of Clinical Pharmacy, Department of Pharmacy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Yu Xiong
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China; Medical Center for Neurological Disorders, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
| | - Yin Zhang
- Laboratory of Clinical Pharmacy, Department of Pharmacy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Qiuxia Xu
- Laboratory of Clinical Pharmacy, Department of Pharmacy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Lingxing Wang
- Medical Center for Neurological Disorders, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China; Department of Neurology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China.
| | - Shaobin He
- Laboratory of Clinical Pharmacy, Department of Pharmacy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China; Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Department of Pharmaceutical Analysis, Fujian Medical University, Fuzhou, 350004, China; Department of Pulmonary and Critical Care Medicine, Fujian Key Laboratory of Lung Stem Cell, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China.
| | - Feng Zheng
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China; Medical Center for Neurological Disorders, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China.
| |
Collapse
|
134
|
Yin L, Xu Y, Mu J, Leng Y, Ma L, Zheng Y, Li R, Wang Y, Li P, Zhu H, Wang D, Li J. CNKSR2 interactome analysis indicates its association with the centrosome/microtubule system. Neural Regen Res 2025; 20:2420-2432. [PMID: 39359098 PMCID: PMC11759008 DOI: 10.4103/nrr.nrr-d-23-01725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/27/2024] [Accepted: 03/04/2024] [Indexed: 10/04/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202508000-00031/figure1/v/2024-09-30T120553Z/r/image-tiff The protein connector enhancer of kinase suppressor of Ras 2 (CNKSR2), present in both the postsynaptic density and cytoplasm of neurons, is a scaffolding protein with several protein-binding domains. Variants of the CNKSR2 gene have been implicated in neurodevelopmental disorders, particularly intellectual disability, although the precise mechanism involved has not yet been fully understood. Research has demonstrated that CNKSR2 plays a role in facilitating the localization of postsynaptic density protein complexes to the membrane, thereby influencing synaptic signaling and the morphogenesis of dendritic spines. However, the function of CNKSR2 in the cytoplasm remains to be elucidated. In this study, we used immunoprecipitation and high-resolution liquid chromatography-mass spectrometry to identify the interactors of CNKSR2. Through a combination of bioinformatic analysis and cytological experiments, we found that the CNKSR2 interactors were significantly enriched in the proteome of the centrosome. We also showed that CNKSR2 interacted with the microtubule protein DYNC1H1 and with the centrosome marker CEP290. Subsequent colocalization analysis confirmed the centrosomal localization of CNKSR2. When we downregulated CNKSR2 expression in mouse neuroblastoma cells (Neuro 2A), we observed significant changes in the expression of numerous centrosomal genes. This manipulation also affected centrosome-related functions, including cell size and shape, cell proliferation, and motility. Furthermore, we found that CNKSR2 interactors were highly enriched in de novo variants associated with intellectual disability and autism spectrum disorder. Our findings establish a connection between CNKSR2 and the centrosome, and offer new insights into the underlying mechanisms of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Lin Yin
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, Shandong Province, China
- School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Yalan Xu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, Shandong Province, China
- School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Jie Mu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, Shandong Province, China
- College of Life Sciences, and School of Pharmacy, Medical College, Qingdao University, Qingdao, Shandong Province, China
| | - Yu Leng
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, Shandong Province, China
- School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Lei Ma
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, Shandong Province, China
- School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Yu Zheng
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, Shandong Province, China
- School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China
- Department of Urology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, Shandong Province, China
| | - Ruizhi Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, Shandong Province, China
- School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, Shandong Province, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, Shandong Province, China
| | - Hai Zhu
- Department of Urology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, Shandong Province, China
| | - Dong Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, Shandong Province, China
| | - Jing Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
135
|
Song R, Yin S, Wu J, Yan J. Neuronal regulated cell death in aging-related neurodegenerative diseases: key pathways and therapeutic potentials. Neural Regen Res 2025; 20:2245-2263. [PMID: 39104166 PMCID: PMC11759035 DOI: 10.4103/nrr.nrr-d-24-00025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 05/13/2024] [Accepted: 06/18/2024] [Indexed: 08/07/2024] Open
Abstract
Regulated cell death (such as apoptosis, necroptosis, pyroptosis, autophagy, cuproptosis, ferroptosis, disulfidptosis) involves complex signaling pathways and molecular effectors, and has been proven to be an important regulatory mechanism for regulating neuronal aging and death. However, excessive activation of regulated cell death may lead to the progression of aging-related diseases. This review summarizes recent advances in the understanding of seven forms of regulated cell death in age-related diseases. Notably, the newly identified ferroptosis and cuproptosis have been implicated in the risk of cognitive impairment and neurodegenerative diseases. These forms of cell death exacerbate disease progression by promoting inflammation, oxidative stress, and pathological protein aggregation. The review also provides an overview of key signaling pathways and crosstalk mechanisms among these regulated cell death forms, with a focus on ferroptosis, cuproptosis, and disulfidptosis. For instance, FDX1 directly induces cuproptosis by regulating copper ion valency and dihydrolipoamide S-acetyltransferase aggregation, while copper mediates glutathione peroxidase 4 degradation, enhancing ferroptosis sensitivity. Additionally, inhibiting the Xc- transport system to prevent ferroptosis can increase disulfide formation and shift the NADP + /NADPH ratio, transitioning ferroptosis to disulfidptosis. These insights help to uncover the potential connections among these novel regulated cell death forms and differentiate them from traditional regulated cell death mechanisms. In conclusion, identifying key targets and their crosstalk points among various regulated cell death pathways may aid in developing specific biomarkers to reverse the aging clock and treat age-related neurodegenerative conditions.
Collapse
Affiliation(s)
- Run Song
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Shiyi Yin
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Jiannan Wu
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Junqiang Yan
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| |
Collapse
|
136
|
Mori K, Togo A, Yamashita K, Sakuragi S, Bannai H, Umezawa T, Ohta K, Asahi T, Nozaki C, Kataoka K. Mitochondrial damage and ER stress in CB1 receptor antagonist-induced apoptosis in human neuroblastoma SH-SY5Y cells. Neuropharmacology 2025; 273:110440. [PMID: 40185361 DOI: 10.1016/j.neuropharm.2025.110440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/13/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
Cannabinoid receptor type 1 (CB1R) is the key modulator of neuronal viability. CB1R antagonists provide neuroprotective effects on neurotoxicity caused by e.g. neuronal injury. However, the underlying mechanisms and potential limitations of CB1R antagonism remain unclear. Here we investigated the impact of environmental conditions on CB1R antagonist effects. We have found that cell-permeable CB1R antagonists, rimonabant and AM251, induced cell death in human neuroblastoma SH-SY5Y cells under serum-free conditions. Mitochondrial morphological analysis revealed mitochondrial swelling characterized by their network fragmentation and cristae reduction. Phosphoproteomics analysis showed the ER stress signaling pathway PERK/eIF2α/ATF4/CHOP, leading to caspase-dependent apoptosis. These results suggest that CB1R antagonists promote apoptosis via mitochondrial damage and ER stress under serum-free conditions in SH-SY5Y cells. Our findings indicate that while CB1R antagonists may be neuroprotective in certain conditions, they may also pose a neurotoxic risk in environments characterized by cellular stress or nutrient deprivation.
Collapse
Affiliation(s)
- Kazuaki Mori
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Akinobu Togo
- Advanced Imaging Research Center, Kurume University School of Medicine, 67 Asahi-cho, Kurume-shi, Fukuoka, 830-0011, Japan
| | - Kota Yamashita
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei-shi, Tokyo, 184-8588, Japan
| | - Shigeo Sakuragi
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Hiroko Bannai
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Taishi Umezawa
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei-shi, Tokyo, 184-8588, Japan
| | - Keisuke Ohta
- Advanced Imaging Research Center, Kurume University School of Medicine, 67 Asahi-cho, Kurume-shi, Fukuoka, 830-0011, Japan
| | - Toru Asahi
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan; Comprehensive Research Organization, Waseda University, 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo, 162-0041, Japan
| | - Chihiro Nozaki
- Global Center for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Kosuke Kataoka
- Comprehensive Research Organization, Waseda University, 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo, 162-0041, Japan; Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei-shi, Tokyo, 184-8588, Japan.
| |
Collapse
|
137
|
Wallace CW, Holleran KM, Slinkard CY, Centanni SW, Lapish CC, Jones SR. Kappa opioid receptors diminish spontaneous dopamine signals in awake mice through multiple mechanisms. Neuropharmacology 2025; 273:110458. [PMID: 40204058 DOI: 10.1016/j.neuropharm.2025.110458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 03/06/2025] [Accepted: 04/03/2025] [Indexed: 04/11/2025]
Abstract
The role of the dynorphin/kappa opioid receptor (KOR) system in dopamine (DA) regulation has been extensively investigated. KOR activation reduces extracellular DA concentrations, but the exact mechanism(s) through which this is accomplished are not fully elucidated. To explore KOR influences on real-time DA fluctuations, we used the photosensor dLight1.2 with fiber photometry in the nucleus accumbens (NAc) core of freely moving male and female C57BL/6J mice. First, we established that the rise and fall of spontaneously arising DA signals were due to DA release and reuptake, respectively. Next, mice were systemically administered the KOR agonist U50,488H in the presence or absence of the KOR antagonist aticaprant. U50,488H reduced both the amplitude and width of spontaneous signals in both sexes. Further, the slope of the correlation between amplitude and width was increased, indicating that DA uptake rates were increased. U50,488H also reduced the frequency of occurrence of signals in males and females. The effects of KOR activation were stronger in males, while effects of KOR antagonism were stronger in females. Overall, KORs exerted significant inhibitory control over spontaneous DA signaling, acting through at least three mechanisms - inhibiting DA release, promoting DA transporter-mediated uptake, and reducing the frequency of signals.
Collapse
Affiliation(s)
- Conner W Wallace
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Katherine M Holleran
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Clare Y Slinkard
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Samuel W Centanni
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Christopher C Lapish
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sara R Jones
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
138
|
Periyasamy TS, Kasivishwanathan A, Roy G, Sekar N, Lakshmanan H. Phytocompounds of Senecio candicans as potential acetylcholinesterase inhibitors targeting Alzheimer's disease: A structure-based virtual screening and molecular dynamics simulation study. Comput Biol Chem 2025; 117:108396. [PMID: 40024051 DOI: 10.1016/j.compbiolchem.2025.108396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/04/2025]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder characterized by cognitive decline due to the accumulation of amyloid-beta plaques, neurofibrillary tangles, and decreased acetylcholine levels caused by acetylcholinesterase (AChE) activity. Current treatments using synthetic acetylcholinesterase inhibitors (AChEIs) provide only symptomatic relief and are associated with adverse effects, highlighting the need for safer and more effective alternatives. This study investigates the potential of phytoconstituents from the plant Senecio candicans as natural AChE inhibitors for AD treatment. Using structure-based virtual screening, molecular docking, and molecular dynamics simulations, we evaluated several compounds from Senecio candicans for their binding affinity, stability, and inhibitory activity against AChE. The findings identified compounds such as Estra-135(10)-trien-17β-ol and Vulgarone A, which demonstrated strong binding affinities and stable interactions with AChE, comparable to or surpassing the clinically used drug Donepezil. These phytoconstituents exhibited potential as effective AChEIs with potentially fewer side effects. The results underscore the therapeutic potential of plant-based molecules for drug discovery, offering a promising avenue for developing new treatments for neurodegenerative diseases. Combining phytochemical studies with computational methods provides a powerful approach to identifying novel therapeutic agents. This study suggests that phytoconstituents from Senecio candicans could serve as safer alternatives for managing AD. Further experimental validation and clinical studies are necessary to confirm these compounds' efficacy and safety, paving the way for innovative, plant-derived treatments for Alzheimer's disease.
Collapse
Affiliation(s)
- Tamilarasi Sambu Periyasamy
- Division of Biochemistry, School of Life Sciences, Ooty campus, JSS Academy of Higher Education and Research, Mysuru road, Longwood, Ooty, The Nilgiris, Tamil Nadu, India
| | - Ajay Kasivishwanathan
- Division of Biochemistry, School of Life Sciences, Ooty campus, JSS Academy of Higher Education and Research, Mysuru road, Longwood, Ooty, The Nilgiris, Tamil Nadu, India
| | - Gilbert Roy
- Division of Biochemistry, School of Life Sciences, Ooty campus, JSS Academy of Higher Education and Research, Mysuru road, Longwood, Ooty, The Nilgiris, Tamil Nadu, India
| | - Nishu Sekar
- Division of Biochemistry, School of Life Sciences, Ooty campus, JSS Academy of Higher Education and Research, Mysuru road, Longwood, Ooty, The Nilgiris, Tamil Nadu, India
| | - Hariprasath Lakshmanan
- Division of Biochemistry, School of Life Sciences, Ooty campus, JSS Academy of Higher Education and Research, Mysuru road, Longwood, Ooty, The Nilgiris, Tamil Nadu, India.
| |
Collapse
|
139
|
Norte-Muñoz M, Portela-Lomba M, Sobrado-Calvo P, Simón D, Di Pierdomenico J, Gallego-Ortega A, Pérez M, Cabrera-Maqueda JM, Sierra J, Vidal-Sanz M, Moreno-Flores MT, Agudo-Barriuso M. Differential response of injured and healthy retinas to syngeneic and allogeneic transplantation of a clonal cell line of immortalized olfactory ensheathing glia: a double-edged sword. Neural Regen Res 2025; 20:2395-2407. [PMID: 39359096 PMCID: PMC11759016 DOI: 10.4103/nrr.nrr-d-23-01631] [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: 09/28/2023] [Revised: 12/01/2023] [Accepted: 04/19/2024] [Indexed: 10/04/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202508000-00029/figure1/v/2024-09-30T120553Z/r/image-tiff Olfactory ensheathing glia promote axonal regeneration in the mammalian central nervous system, including retinal ganglion cell axonal growth through the injured optic nerve. Still, it is unknown whether olfactory ensheathing glia also have neuroprotective properties. Olfactory ensheathing glia express brain-derived neurotrophic factor, one of the best neuroprotectants for axotomized retinal ganglion cells. Therefore, we aimed to investigate the neuroprotective capacity of olfactory ensheating glia after optic nerve crush. Olfactory ensheathing glia cells from an established rat immortalized clonal cell line, TEG3, were intravitreally injected in intact and axotomized retinas in syngeneic and allogeneic mode with or without microglial inhibition or immunosuppressive treatments. Anatomical and gene expression analyses were performed. Olfactory bulb-derived primary olfactory ensheathing glia and TEG3 express major histocompatibility complex class II molecules. Allogeneically and syngenically transplanted TEG3 cells survived in the vitreous for up to 21 days, forming an epimembrane. In axotomized retinas, only the allogeneic TEG3 transplant rescued retinal ganglion cells at 7 days but not at 21 days. In these retinas, microglial anatomical activation was higher than after optic nerve crush alone. In intact retinas, both transplants activated microglial cells and caused retinal ganglion cell death at 21 days, a loss that was higher after allotransplantation, triggered by pyroptosis and partially rescued by microglial inhibition or immunosuppression. However, neuroprotection of axotomized retinal ganglion cells did not improve with these treatments. The different neuroprotective properties, different toxic effects, and different responses to microglial inhibitory treatments of olfactory ensheathing glia in the retina depending on the type of transplant highlight the importance of thorough preclinical studies to explore these variables.
Collapse
Affiliation(s)
- María Norte-Muñoz
- Grupo de Investigación Oftalmología Experimental, Departamento de Oftalmología, Optometría, Otorrinolaringología y Anatomía Patológica, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Campus de Ciencias de la Salud, Murcia, Spain
| | - María Portela-Lomba
- Experimental Sciences Faculty, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - Paloma Sobrado-Calvo
- Grupo de Investigación Oftalmología Experimental, Departamento de Oftalmología, Optometría, Otorrinolaringología y Anatomía Patológica, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Campus de Ciencias de la Salud, Murcia, Spain
| | - Diana Simón
- Experimental Sciences Faculty, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - Johnny Di Pierdomenico
- Grupo de Investigación Oftalmología Experimental, Departamento de Oftalmología, Optometría, Otorrinolaringología y Anatomía Patológica, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Campus de Ciencias de la Salud, Murcia, Spain
| | - Alejandro Gallego-Ortega
- Grupo de Investigación Oftalmología Experimental, Departamento de Oftalmología, Optometría, Otorrinolaringología y Anatomía Patológica, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Campus de Ciencias de la Salud, Murcia, Spain
| | - Mar Pérez
- Anatomy, Histology and Neuroscience Department, Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
| | - José M. Cabrera-Maqueda
- Grupo de Investigación Oftalmología Experimental, Departamento de Oftalmología, Optometría, Otorrinolaringología y Anatomía Patológica, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Campus de Ciencias de la Salud, Murcia, Spain
- Center of Neuroimmunology, Service of Neurology, Laboratory of Advanced Imaging in Neuroimmunological Diseases, Hospital Clínic of Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Universitat de Barcelona, Barcelona, Spain
| | - Javier Sierra
- Medicine Faculty, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - Manuel Vidal-Sanz
- Grupo de Investigación Oftalmología Experimental, Departamento de Oftalmología, Optometría, Otorrinolaringología y Anatomía Patológica, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Campus de Ciencias de la Salud, Murcia, Spain
| | - María Teresa Moreno-Flores
- Anatomy, Histology and Neuroscience Department, Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
| | - Marta Agudo-Barriuso
- Grupo de Investigación Oftalmología Experimental, Departamento de Oftalmología, Optometría, Otorrinolaringología y Anatomía Patológica, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Campus de Ciencias de la Salud, Murcia, Spain
| |
Collapse
|
140
|
Chen J, Li Y, Quan X, Chen J, Han Y, Yang L, Zhou M, Mok GSP, Wang R, Zhao Y. Utilizing engineered extracellular vesicles as delivery vectors in the management of ischemic stroke: a special outlook on mitochondrial delivery. Neural Regen Res 2025; 20:2181-2198. [PMID: 39101653 PMCID: PMC11759020 DOI: 10.4103/nrr.nrr-d-24-00243] [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: 02/28/2024] [Revised: 06/03/2024] [Accepted: 06/22/2024] [Indexed: 08/06/2024] Open
Abstract
Ischemic stroke is a secondary cause of mortality worldwide, imposing considerable medical and economic burdens on society. Extracellular vesicles, serving as natural nano-carriers for drug delivery, exhibit excellent biocompatibility in vivo and have significant advantages in the management of ischemic stroke. However, the uncertain distribution and rapid clearance of extracellular vesicles impede their delivery efficiency. By utilizing membrane decoration or by encapsulating therapeutic cargo within extracellular vesicles, their delivery efficacy may be greatly improved. Furthermore, previous studies have indicated that microvesicles, a subset of large-sized extracellular vesicles, can transport mitochondria to neighboring cells, thereby aiding in the restoration of mitochondrial function post-ischemic stroke. Small extracellular vesicles have also demonstrated the capability to transfer mitochondrial components, such as proteins or deoxyribonucleic acid, or their sub-components, for extracellular vesicle-based ischemic stroke therapy. In this review, we undertake a comparative analysis of the isolation techniques employed for extracellular vesicles and present an overview of the current dominant extracellular vesicle modification methodologies. Given the complex facets of treating ischemic stroke, we also delineate various extracellular vesicle modification approaches which are suited to different facets of the treatment process. Moreover, given the burgeoning interest in mitochondrial delivery, we delved into the feasibility and existing research findings on the transportation of mitochondrial fractions or intact mitochondria through small extracellular vesicles and microvesicles to offer a fresh perspective on ischemic stroke therapy.
Collapse
Affiliation(s)
- Jiali Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Yiyang Li
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Xingping Quan
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Jinfen Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Yan Han
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Li Yang
- Department of Pharmacy, Hunan Provincial People’s Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Manfei Zhou
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Greta Seng Peng Mok
- Department of Electrical and Computer Engineering, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Ruibing Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Yonghua Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao Special Administrative Region, China
| |
Collapse
|
141
|
Sasaki LMP, Canellas-de-Castro ME, Fernandes GM, Motta F, Araújo Júnior DA, Schulte HL, Jardim-Santos GP, Silva ÂP, Carvalho AO, Pereira YR, Siracusa CC, Rabelo IP, Santos Junior ADCM, Alves CDO, Lauand L, Nery RDR, Mendonça-Silva DL, Tristão R, Jesus JAL, Costa KN, Castro LCG, Kurizky PS, Nóbrega ODT, Espindola LS, Coelho-Dos-Reis JGA, Brito-de-Sousa JP, Costa-Rocha IA, Campi-Azevedo AC, Peruhype-Magalhães V, Teixeira-Carvalho A, Gomes CM, Albuquerque CP, Soares AADSM, Zaconeta AM, Martins-Filho OA, Mota LMH. Systemic and cerebrospinal fluid immune mediators coordinate a dichotomic microenvironment in parturients with acute or convalescent phases of COVID-19. Immunol Lett 2025; 274:106979. [PMID: 39933603 DOI: 10.1016/j.imlet.2025.106979] [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: 11/10/2024] [Revised: 01/17/2025] [Accepted: 02/06/2025] [Indexed: 02/13/2025]
Abstract
The present study intended to characterize the profile of soluble immune mediators in serum samples and in the cerebrospinal fluid (CSF) microenvironment from parturients with acute and convalescent COVID-19 as compared to healthy controls (HC), during the circulation of B.1.1.28 and B.1.1.33 SARS-CoV-2 strains, which were identified during the initial spread of COVID-19 in Brazil. Data demonstrated increased levels of immune mediators in serum at acute infection with a clear waning during convalescent COVID-19. Conversely, a progressive increase of immune mediators was observed in CSF from acute infection towards convalescent COVID-19. Immune mediator signatures and integrative correlation circuits further confirmed these findings and supported the existence of dichotomic microenvironments in the serum and CSF compartments. While a waning of correlations involving pro-inflammatory cytokines with increased connectivity of regulatory cytokines was observed in serum samples from acute towards convalescent COVID-19, an increasing frequency of correlations mediated by pro-inflammatory cytokines with decreased connectivity of regulatory cytokine was the hallmark of CSF. Correlations analysis identified a set of molecules associated with the dichotomic crosstalk between serum and CSF compartments, including chemokines (CXCL8, CCL5, CXCL10) and regulatory cytokines (IL-4 and IL-9). These immune biomarkers may represent potential targets for therapeutic strategies in parturients with COVID-19. Together, these findings demonstrated the existence of a divergent landscape of soluble immune mediators in serum and CSF, emphasizing the relevance of understanding the systemic and compartmentalized immune response elicited by SARS-CoV-2 infection during pregnancy.
Collapse
Affiliation(s)
- Lizandra Moura Paravidine Sasaki
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil; Hospital Universitário de Brasília, Universidade de Brasília (UnB), Brasília, Brazil.
| | - Maria Eduarda Canellas-de-Castro
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil; Hospital Universitário de Brasília, Universidade de Brasília (UnB), Brasília, Brazil; Faculdade de Medicina, Universidade de Brasília (UnB), Brasília, Brazil
| | - Geraldo Magela Fernandes
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil; Faculdade de Medicina, Universidade de Brasília (UnB), Brasília, Brazil
| | - Felipe Motta
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil
| | | | - Heidi Luise Schulte
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil
| | | | - Ângelo Pereira Silva
- Hospital Universitário de Brasília, Universidade de Brasília (UnB), Brasília, Brazil
| | | | | | | | | | | | | | - Lucas Lauand
- Faculdade de Medicina, Universidade de Brasília (UnB), Brasília, Brazil
| | | | | | - Rosana Tristão
- Faculdade de Medicina, Universidade de Brasília (UnB), Brasília, Brazil
| | | | - Karina Nascimento Costa
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil; Faculdade de Medicina, Universidade de Brasília (UnB), Brasília, Brazil
| | - Luiz Cláudio Gonçalves Castro
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil; Faculdade de Medicina, Universidade de Brasília (UnB), Brasília, Brazil
| | - Patricia Shu Kurizky
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil; Hospital Universitário de Brasília, Universidade de Brasília (UnB), Brasília, Brazil
| | | | - Laila Salmen Espindola
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil
| | | | | | | | | | | | | | - Ciro Martins Gomes
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil; Faculdade de Medicina, Universidade de Brasília (UnB), Brasília, Brazil; Programa de Pós-Graduação em Patologia Molecular, Universidade de Brasília (UnB), Brasília, Brazil
| | - Cleandro Pires Albuquerque
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil; Hospital Universitário de Brasília, Universidade de Brasília (UnB), Brasília, Brazil
| | - Alexandre Anderson de Sousa Munhoz Soares
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil; Faculdade de Medicina, Universidade de Brasília (UnB), Brasília, Brazil
| | | | | | - Licia Maria Henrique Mota
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil; Hospital Universitário de Brasília, Universidade de Brasília (UnB), Brasília, Brazil; Programa de Pós-Graduação em Patologia Molecular, Universidade de Brasília (UnB), Brasília, Brazil
| |
Collapse
|
142
|
Yang YT, Romero-Leguizamón CR, Sheykhzade M, Zhu Y, Kohlmeier KA. Calcitonin gene-related peptide (CGRP) exerts membrane, cellular and synaptic actions on serotonergic dorsal raphe neurons ex vivo: Functional implications for a role in dorsal raphe-controlled functions. Neuropharmacology 2025; 273:110457. [PMID: 40189018 DOI: 10.1016/j.neuropharm.2025.110457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Abstract
Serotonin (5-HT) plays a role in limbic-controlled behaviors and is implicated in migraine, which is often co-morbid with cognitive-based affective disorders. The neuropeptide calcitonin gene-related peptide (CGRP) regulates vascular tone. Serotonin-acting drugs and CGRP receptor antagonists have proved therapeutic in management of migraine. Clinical interactions between the two systems have been shown, however, whether CGRP exerts direct actions on serotonergic Dorsal Raphe (DR) neurons is unknown. To fully understand the role of CGRP in control of behavior and to predict how CGRP targeted therapies (i.e. CGRP receptor antagonists) could alter DR neuronal activity, investigation of whether CGRP can directly affect 5-HT DR activity was conducted. Patch clamp electrophysiology and single photon calcium imaging in DR brain slices revealed that CGRP (10-6 M) elicited postsynaptically mediated, potassium-involved outward currents in the majority of 5-HT DR cells. Miniature excitatory synaptic events were reduced in frequency. Further, intracellular calcium was reduced in the majority of neurons, which did not involve actions on the L-type calcium channel. The CGRP agonist SAX replicated effects on the membrane and intracellular calcium. In contrast, the CGRP receptor antagonist MK-3207 blocked the effects on outward current and attenuated the action of CGRP on reducing intracellular calcium. Despite inhibitory membrane and synaptic effects, no change was noted in firing rate. Our findings raise the intriguing possibility that the CGRP system plays a role in mediating limbic-controlled behaviors, at least in part, through direct actions on serotonergic DR neurons, however the effect of CGRP on DR 5-HT output remains to be investigated.
Collapse
Affiliation(s)
- Yan Tong Yang
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, 2100, Denmark; Sino-Danish Center for Education and Research, Beijing, 101408, China; State Key Laboratory of Cognitive Science and Mental Health, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing, 100101, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 101408, China; Jinfeng Laboratory, Chongqing 401329, China
| | | | - Majid Sheykhzade
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Yan Zhu
- Sino-Danish Center for Education and Research, Beijing, 101408, China; State Key Laboratory of Cognitive Science and Mental Health, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing, 100101, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 101408, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kristi Anne Kohlmeier
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, 2100, Denmark; Sino-Danish Center for Education and Research, Beijing, 101408, China.
| |
Collapse
|
143
|
Wu Y, Yang L, Jiang W, Zhang X, Yao Z. Glycolytic dysregulation in Alzheimer's disease: unveiling new avenues for understanding pathogenesis and improving therapy. Neural Regen Res 2025; 20:2264-2278. [PMID: 39101629 PMCID: PMC11759019 DOI: 10.4103/nrr.nrr-d-24-00190] [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: 02/14/2024] [Revised: 05/11/2024] [Accepted: 06/20/2024] [Indexed: 08/06/2024] Open
Abstract
Alzheimer's disease poses a significant global health challenge owing to the progressive cognitive decline of patients and absence of curative treatments. The current therapeutic strategies, primarily based on cholinesterase inhibitors and N-methyl-D-aspartate receptor antagonists, offer limited symptomatic relief without halting disease progression, highlighting an urgent need for novel research directions that address the key mechanisms underlying Alzheimer's disease. Recent studies have provided insights into the critical role of glycolysis, a fundamental energy metabolism pathway in the brain, in the pathogenesis of Alzheimer's disease. Alterations in glycolytic processes within neurons and glial cells, including microglia, astrocytes, and oligodendrocytes, have been identified as significant contributors to the pathological landscape of Alzheimer's disease. Glycolytic changes impact neuronal health and function, thus offering promising targets for therapeutic intervention. The purpose of this review is to consolidate current knowledge on the modifications in glycolysis associated with Alzheimer's disease and explore the mechanisms by which these abnormalities contribute to disease onset and progression. Comprehensive focus on the pathways through which glycolytic dysfunction influences Alzheimer's disease pathology should provide insights into potential therapeutic targets and strategies that pave the way for groundbreaking treatments, emphasizing the importance of understanding metabolic processes in the quest for clarification and management of Alzheimer's disease.
Collapse
Affiliation(s)
- You Wu
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Lijie Yang
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Wanrong Jiang
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Xinyuan Zhang
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Zhaohui Yao
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
144
|
Morín IO, Depaepe F, Reynvoet B. Sharpening the number sense: Developmental trends in numerosity perception. J Exp Child Psychol 2025; 256:106262. [PMID: 40239424 DOI: 10.1016/j.jecp.2025.106262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 03/21/2025] [Accepted: 03/23/2025] [Indexed: 04/18/2025]
Abstract
Numerosity perception, the ability to process and estimate the number of objects in a set without explicitly counting, has been widely studied, and one well-established finding is that children become more accurate at perceiving numerosity with age. The question remains, however, what the underlying cognitive processes and mechanisms are that drive this improvement. Some authors have suggested that this is due to an increased numerical precision (i.e., the sharpening hypothesis), whereas others have proposed that the more accurate performance is due to the improved ability to inhibit non-numerical features of the display such as object size and spacing of items (i.e., the filtering hypothesis). The current study examined the developmental trajectory of numerosity perception across three age groups (M = 5.65, M = 11.03, and M = 20.10 years). As expected, more accurate performance was observed with age. Regression and analyses of variance revealing the contribution of numerical and non-numerical predictors in performance show that the performance in all age groups was primarily driven by numerical information and that its contribution increased with age. In addition, a consistent bias toward non-numerical features was observed in all age groups. These results support the sharpening hypothesis for children from 5 years of age to early adulthood, suggesting that from this age onward children increasingly focus on numerical information as they get older. These results have important implications for the understanding of the development and specific improvements of numerical perception.
Collapse
Affiliation(s)
- Irene Oeo Morín
- Brain and Cognition, KU (Katholieke Universiteit) Leuven 3000 Leuven, Belgium; Faculty of Psychology and Educational Sciences, KU Leuven, Kulak Campus, 8500 Kortrijk, Belgium.
| | - Fien Depaepe
- Faculty of Psychology and Educational Sciences, KU Leuven, Kulak Campus, 8500 Kortrijk, Belgium; ITEC, IMEC (Interuniversity Microelectronics Centre) Research Group, KU Leuven, Kulak Campus, 8500 Kortrijk, Belgium
| | - Bert Reynvoet
- Brain and Cognition, KU (Katholieke Universiteit) Leuven 3000 Leuven, Belgium; Faculty of Psychology and Educational Sciences, KU Leuven, Kulak Campus, 8500 Kortrijk, Belgium
| |
Collapse
|
145
|
Su S, Levasseur IN, Alonge KM. Neurocircuit regeneration by extracellular matrix reprogramming. Neural Regen Res 2025; 20:2300-2301. [PMID: 39359079 PMCID: PMC11759010 DOI: 10.4103/nrr.nrr-d-24-00506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 06/22/2024] [Accepted: 07/09/2024] [Indexed: 10/04/2024] Open
Affiliation(s)
- Shengzhang Su
- Department of Chemistry, University of Washington, Seattle, WA, USA
| | - Ian N. Levasseur
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Kimberly M. Alonge
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington Medicine Diabetes Institute, Seattle, WA, USA
| |
Collapse
|
146
|
Izumi S, Kawasaki I, Waki F, Nishikawa K, Nishitani N, Deyama S, Kaneda K. Chronic nicotine enhances object recognition memory via inducing long-term potentiation in the medial prefrontal cortex in mice. Neuropharmacology 2025; 273:110435. [PMID: 40154943 DOI: 10.1016/j.neuropharm.2025.110435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Chronic nicotine administration enhances cognitive functions, including learning and memory, and ameliorates cognitive impairments observed in psychological and neurodegenerative disorders. However, the detailed mechanisms underlying these effects are not fully understood. In this study, we used a novel object recognition (NOR) test and in vitro slice electrophysiology in mice to investigate the involvement of the medial prefrontal cortex (mPFC), a brain region connected to the hippocampus, and the synaptic plasticity within this region in chronic nicotine-induced object recognition memory enhancement. The NOR test revealed that chronic nicotine administration for five consecutive days significantly enhanced object recognition memory in male and female mice. This effect was blocked by intra-mPFC infusion of mecamylamine (Mec), a non-selective nicotinic acetylcholine receptor (nAChR) antagonist. In parallel with these findings, whole-cell recordings demonstrated that chronic nicotine administration significantly increased the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)/N-methyl-d-aspartate (NMDA) ratio in mPFC layer V pyramidal neurons in male but not female mice. This plastic change was suppressed by systemic injection of Mec or methyllycaconitine, an α7 nAChR antagonist. Furthermore, optogenetic erasure of long-term potentiation (LTP) through chromophore-assisted light inactivation of cofilin, a protein essential for stabilizing spine expansion, suppressed chronic nicotine-induced enhancement of recognition memory. These findings suggest that chronic nicotine administration induces LTP in mPFC pyramidal neurons, likely enhancing object recognition memory.
Collapse
Affiliation(s)
- Shoma Izumi
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Ibuki Kawasaki
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Fuka Waki
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Keisuke Nishikawa
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Naoya Nishitani
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Satoshi Deyama
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Katsuyuki Kaneda
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan.
| |
Collapse
|
147
|
Rosell-Cardona C, Collins MK, O'Riordan KJ, Goodson MS, Kelley-Loughnane N, Cryan JF, Clarke G. Acute stress enhances synaptic plasticity in male mice via a microbiota-dependent mechanism. Neuropharmacology 2025; 273:110434. [PMID: 40154944 DOI: 10.1016/j.neuropharm.2025.110434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/25/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Acute stress can enhance or impair synaptic plasticity depending on the nature, duration, and type of stress exposure as well as the brain region examined. The absence of a gut microbiome can also alter hippocampal plasticity. However, the possible interplay between synaptic plasticity, acute stress, and the gut microbiota remains unknown. Here, we examine this interaction and determine whether the gut microbiota impacts stress-induced alterations in hippocampal plasticity. Further, we explored whether exposure to the microbial metabolite butyrate is sufficient to counteract stress-induced alterations in synaptic plasticity. We used electrophysiological and molecular experiments in adult male C57/BL6 antibiotic-treated and acutely stressed mice. In electrophysiological experiments we treated hippocampal slices with 3 μM sodium butyrate to explore the effect of this microbial metabolite. We found the presence of the microbiota essential for the enhancement of both short- and long-term potentiation induced by 15 min of acute restraint stress. Furthermore, butyrate exposure effectively restored the stress-induced enhancement of potentiation in slices from microbiome-depleted animals while also enhancing long-term potentiation independent of stress. In addition, alterations of hippocampal synaptic plasticity markers were noted. Our findings highlight a critical new temporal role for gut-derived metabolites in defining the impact of acute stress on synaptic plasticity.
Collapse
Affiliation(s)
| | - Michael K Collins
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | | | - Michael S Goodson
- 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson Air Force Base, Dayton, OH, USA
| | - Nancy Kelley-Loughnane
- 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson Air Force Base, Dayton, OH, USA
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland.
| |
Collapse
|
148
|
Boulksibat A, Tempio A, Bardoni B. Central role of altered phosphodiesterase 2-dependent signaling in the pathophysiology of cognition-based brain disorders. Neural Regen Res 2025; 20:2302-2303. [PMID: 39359080 PMCID: PMC11759023 DOI: 10.4103/nrr.nrr-d-24-00588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/13/2024] [Accepted: 06/27/2024] [Indexed: 10/04/2024] Open
Affiliation(s)
- Asma Boulksibat
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR7275, Inserm U1323, Valbonne, France
| | - Alessandra Tempio
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR7275, Inserm U1323, Valbonne, France
| | - Barbara Bardoni
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR7275, Inserm U1323, Valbonne, France
| |
Collapse
|
149
|
Huang L, Wang Y, Sun X, Deng K, Li X, Xie Y, Guo H, Zhao P, Fei J. Square-shaped Cu 2MoS 4 loaded on three-dimensional flower-like AgBiS 2 to form S-scheme heterojunction as a light-driven photoelectrochemical sensor for efficient detection of serotonin in biological samples. Talanta 2025; 290:127774. [PMID: 40015067 DOI: 10.1016/j.talanta.2025.127774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/27/2025] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
Serotonin (5-HT) is a crucial neurotransmitter in the body, with its levels being particularly significant for life safety. Here, we designed the AgBiS2/Cu2MoS4 S-scheme heterojunction by uniformly immobilizing lamellar Cu2MoS4 on the surface of three-dimensional (3D) flower-like AgBiS2 using a simple physical mixing technique. In this case, AgBiS2 and Cu2MoS4 are bonded together by electrostatic attraction to form an active surface with a large specific surface area. Subsequently, the detector 5-HT bound to AgBiS2/Cu2MoS4/GCE undergoes hole oxidation and the photocurrent signal increases significantly. Meanwhile, the reaction mechanism of AgBiS2/Cu2MoS4 composite material was investigated through density functional theory calculations. The AgBiS2/Cu2MoS4/GCE sensor demonstrates a low detection limit of 0.046 nM and a wide linear range (0.0001-8 μM). Furthermore, by comparing UV-Vis spectrophotometry and fluorescence spectroscopy for the detection of 5-HT in human serum, it was proved that the sensor has an impressive recovery rate.
Collapse
Affiliation(s)
- Linzi Huang
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, People's Republic of China
| | - Yilin Wang
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, People's Republic of China
| | - Xiaoqian Sun
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, People's Republic of China
| | - Kunxiang Deng
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, People's Republic of China
| | - Xinyi Li
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, People's Republic of China
| | - Yixi Xie
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Xiangtan University, Xiangtan, 411105, People's Republic of China; Hunan Provincial University Key Laboratory for Environmental and Ecological Health, Xiangtan University, Xiangtan, 411105, People's Republic of China
| | - Haoran Guo
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Xiangtan University, Xiangtan, 411105, People's Republic of China.
| | - Pengcheng Zhao
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, People's Republic of China; Hunan Provincial University Key Laboratory for Environmental and Ecological Health, Xiangtan University, Xiangtan, 411105, People's Republic of China.
| | - Junjie Fei
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, People's Republic of China; Hunan Institute of Advanced Sensing and Information Technology, Xiangtan University, Xiangtan, 411105, People's Republic of China.
| |
Collapse
|
150
|
Ma Y, Dong T, Luan F, Yang J, Miao F, Wei P. Interaction of major facilitator superfamily domain containing 2A with the blood-brain barrier. Neural Regen Res 2025; 20:2133-2152. [PMID: 39248155 PMCID: PMC11759009 DOI: 10.4103/nrr.nrr-d-24-00191] [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: 02/15/2024] [Revised: 06/02/2024] [Accepted: 07/08/2024] [Indexed: 09/10/2024] Open
Abstract
The functional and structural integrity of the blood-brain barrier is crucial in maintaining homeostasis in the brain microenvironment; however, the molecular mechanisms underlying the formation and function of the blood-brain barrier remain poorly understood. The major facilitator superfamily domain containing 2A has been identified as a key regulator of blood-brain barrier function. It plays a critical role in promoting and maintaining the formation and functional stability of the blood-brain barrier, in addition to the transport of lipids, such as docosahexaenoic acid, across the blood-brain barrier. Furthermore, an increasing number of studies have suggested that major facilitator superfamily domain containing 2A is involved in the molecular mechanisms of blood-brain barrier dysfunction in a variety of neurological diseases; however, little is known regarding the mechanisms by which major facilitator superfamily domain containing 2A affects the blood-brain barrier. This paper provides a comprehensive and systematic review of the close relationship between major facilitator superfamily domain containing 2A proteins and the blood-brain barrier, including their basic structures and functions, cross-linking between major facilitator superfamily domain containing 2A and the blood-brain barrier, and the in-depth studies on lipid transport and the regulation of blood-brain barrier permeability. This comprehensive systematic review contributes to an in-depth understanding of the important role of major facilitator superfamily domain containing 2A proteins in maintaining the structure and function of the blood-brain barrier and the research progress to date. This will not only help to elucidate the pathogenesis of neurological diseases, improve the accuracy of laboratory diagnosis, and optimize clinical treatment strategies, but it may also play an important role in prognostic monitoring. In addition, the effects of major facilitator superfamily domain containing 2A on blood-brain barrier leakage in various diseases and the research progress on cross-blood-brain barrier drug delivery are summarized. This review may contribute to the development of new approaches for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Yilun Ma
- College of Pharmacy and First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Taiwei Dong
- College of Pharmacy and First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Fei Luan
- College of Pharmacy and First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Juanjuan Yang
- National Drug Clinical Trial Agency, The Second Affiliated Hospital of Shaanxi University of Chinese Medicine/Xixian New District Central Hospital, Xi′an, Shaanxi Province, China
| | - Feng Miao
- College of Pharmacy and First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Peifeng Wei
- National Drug Clinical Trial Agency, The Second Affiliated Hospital of Shaanxi University of Chinese Medicine/Xixian New District Central Hospital, Xi′an, Shaanxi Province, China
| |
Collapse
|