1
|
Smith S, Smith J, Jones K, Castillo A, Wiemann N, Howard-Cunningham A, Cunningham M. Placental ischemia during pregnancy induces hypertension, cerebral inflammation, and oxidative stress in dams postpartum. Hypertens Pregnancy 2025; 44:2454597. [PMID: 39885618 PMCID: PMC11849403 DOI: 10.1080/10641955.2025.2454597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/07/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Preeclampsia (PE) is characterized as de novo hypertension (HTN) with end-organ damage, especially in the brain. PE is hypothesized to be caused by placental ischemia. PE affects ~5-8% of USA pregnancies and increases the risk for HTN and cerebrovascular diseases (CVD) later in life. We hypothesize that blood pressure (BP), cerebral oxidative stress, and cerebral inflammation will increase in postpartum (PP) placental ischemic dams. METHODS Placental ischemia was induced in pregnant Sprague Dawley dams, utilizing reduced uterine perfusion pressure (RUPP) surgery. At 6 weeks PP (~3 human years), BP was measured via carotid catheterization, and cerebral oxidative stress and inflammation were assessed via ELISAs, biochemical assays, and Western blots. RESULTS BP, cerebral pro-inflammatory cytokines (TNF-α and IL-6), and GFAP (a marker of astrocyte activity) were increased in PP RUPP dams. Cerebral hydrogen peroxide (H2O2) was also increased in PP RUPP dams, and had a strong correlation with PP RUPP BP, proinflammatory cytokines (TNF- α and IL-6), and GFAP astrocyte activation. CONCLUSION PP RUPP dams have increased BP, cerebral oxidative stress, and cerebral inflammation at 6 weeks postpartum. These changes in cerebral inflammation and oxidative stress may contribute to the pathology and development of HTN and CVDs in postpartum dams.
Collapse
Affiliation(s)
- Savanna Smith
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX
| | - Jonna Smith
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX
| | - Kylie Jones
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX
| | - Angie Castillo
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX
| | - Natalia Wiemann
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX
| | | | - Mark Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX
| |
Collapse
|
2
|
Zhao Z, Wang R, Ge H, Hou L, Hatano T, Hattori N, Su H, Wang Q, Zhao J. ECHS1-NOX4 interaction suppresses rotenone-induced dopaminergic neurotoxicity through inhibition of mitochondrial ROS production. Free Radic Biol Med 2025; 232:56-71. [PMID: 40032032 DOI: 10.1016/j.freeradbiomed.2025.02.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/16/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Parkinson's disease (PD) is the most common neurodegenerative movement disorder with uncleared mechanisms. Short-chain enoyl-CoA hydratase 1 (ECHS1) is a mitochondrial enzyme critical for the β-oxidation of fatty acids and ATP production. This study aims to explore the roles of ECHS1 in PD by using rotenone-induced experimental PD models. METHODS To evaluate the role of ECHS1 in rotenone-induced dopaminergic neurodegeneration, adeno-associated virus (AAV)-ECHS1 was stereotactically injected into the substantia nigra region of mice to overexpress ECHS1. Motor function of mice among groups was detected by rotarod test and gait analysis. Neurodegeneration, mitochondrial dysfunction and apoptosis were determined by immunohistochemistry, immunofluorescence staining, Western blot or kits, respectively. RESULTS The expression and activity of ECHS1 were decreased in PD mice and positive correlations between ECHS1 reduction and dopaminergic neurodegeneration were observed. Overexpression of ECHS1 by AAV delivery attenuated loss of dopaminergic neuron and motor deficits in PD mice. Mechanistically, ECHS1 attenuated rotenone-induced mitochondrial swelling and loss of cristae as well as decrease of ATP production, mitochondrial membrane potential, complex I/IV activities and oxygen consumption rate (OCR). Mitochondrial ROS (mtROS)-targeted antioxidant mito-TEMPO prevented ECHS1 silence-mediated mitochondrial dysfunction. Furthermore, we found that ECHS1 interacted with NADPH oxidase 4 (NOX4), resulting in decrease of NOX4 activation and subsequent reduction of mtROS production and mitochondrial dysfunction. Finally, inhibition of NOX4 by GLX351322 or mtROS production by mito-TEMPO greatly reduced ECHS1 silence-mediated apoptosis in rotenone-treated SH-SY5Y cells. CONCLUSIONS ECHS1 counteracted dopaminergic neurodegeneration through inhibition of mtROS and restoration of mitochondrial function via interaction with NOX4. Given the central role of mitochondrial dysfunction in PD pathogenesis, elucidating the role of ECHS1 holds great promise for uncovering novel therapeutic targets.
Collapse
Affiliation(s)
- Zirui Zhao
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning Province, 116044, China; School of Integrated Chinese and Western Medicine, Dalian Medical University, Dalian, Liaoning Province, 116044, China
| | - Ruonan Wang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning Province, 116044, China; School of Integrated Chinese and Western Medicine, Dalian Medical University, Dalian, Liaoning Province, 116044, China
| | - Haitao Ge
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning Province, 116044, China
| | - Liyan Hou
- Dalian Medical University Library, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, Liaoning Province, 116044, China
| | - Taku Hatano
- Department of Neurology, Juntendo University Faculty of Medicine. 2-1-1, Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan; Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain Science 2-1 Hirosawa, Wako-city, Saitama, 351-0198, Japan
| | - Hong Su
- School of Health-Preservation and Wellness, Dalian Medical University, Dalian, Liaoning Province, 116044, China
| | - Qingshan Wang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning Province, 116044, China; School of Public Health, Dalian Medical University, Dalian, Liaoning Province, 116044, China.
| | - Jie Zhao
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning Province, 116044, China; School of Integrated Chinese and Western Medicine, Dalian Medical University, Dalian, Liaoning Province, 116044, China.
| |
Collapse
|
3
|
Nisha Aji K, Lalang N, Ramos-Jiménez C, Rahimian R, Mechawar N, Turecki G, Chartrand D, Boileau I, Meyer JH, Rusjan PM, Mizrahi R. Evidence of altered monoamine oxidase B, an astroglia marker, in early psychosis and high-risk state. Mol Psychiatry 2025; 30:2049-2058. [PMID: 39511452 DOI: 10.1038/s41380-024-02816-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 10/22/2024] [Accepted: 10/25/2024] [Indexed: 11/15/2024]
Abstract
A novel radiotracer, [11C]SL25.1188, targets monoamine oxidase-B (MAO-B) enzyme, found primarily in astrocytes, which metabolizes monoamines (including dopamine), particularly in subcortical regions. Altered astrocyte function in schizophrenia is supported by convergent evidence from post-mortem, genetic, transcriptomic, peripheral and preclinical findings. We aimed to test whether levels of MAO-B, an index of astrocyte function are low in the living brains of early psychosis and their high-risk states. Thirty-eight participants including antipsychotic-free/minimally exposed clinical participants with first-episode psychosis (FEP), clinical high-risk (CHR) individuals and healthy volunteers (HVs) underwent a 90-min positron emission tomography (PET) scan with [11C]SL25.1188, to measure MAO-B VT, an index of MAO-B concentration. Participants were excluded if tested positive on urine drug screen (except for cannabis). This study of 14 FEP (mean[SD] age, 25.7[5.7] years; 6 F), 7 CHR (mean[SD] age, 20.9[3.7] years; 4 F) and 17 HV (mean[SD] age, 31.2[13.9] years; 9 F) demonstrated significant group differences in regional MAO-B VT (F(2,37.42) = 4.56, p = 0.02, Cohen's f = 0.49), controlling for tobacco (F (1,37.42) = 5.37, p = 0.03) and cannabis use (F(1,37.42) = 5.11, p = 0.03) with significantly lower MAO-B VT in CHR compared to HV (Cohen's d = 0.99). We report a significant cannabis effect on MAO-B VT (F(1,39.19) = 12.57, p = 0.001, Cohen's f = 0.57), with a significant group-by-cannabis interaction (F(2,37.30) = 3.82, p = 0.03, Cohen's f = 0.45), indicating lower MAO-B VT in cannabis-using clinical groups. Lower MAO-B VT levels were more robust in striatal than cortical regions, in both clinical groups (F(12,46.84) = 2.08, p = 0.04, Cohen's f = 0.73) and in cannabis users (F(6,46.84) = 6.42, p < 0.001, Cohen's f = 0.91). Lower MAO-B concentration supports astrocyte dysfunction in cannabis-using CHR and FEP clinical populations. Lower MAO-B is consistent with replicated striatal dopamine elevation in psychosis, as well as astrocyte dysfunction in schizophrenia.
Collapse
Affiliation(s)
- Kankana Nisha Aji
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
- Clinical and Translational Sciences Lab, Douglas Research Centre, Montreal, QC, Canada
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Nittha Lalang
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
- Vertex Pharmaceuticals, Boston, MA, USA
| | - Christian Ramos-Jiménez
- Clinical and Translational Sciences Lab, Douglas Research Centre, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Reza Rahimian
- Douglas Mental Health University Institute, McGill Group for Suicide Studies, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Naguib Mechawar
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, McGill Group for Suicide Studies, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Gustavo Turecki
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, McGill Group for Suicide Studies, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Daniel Chartrand
- Department of Anesthesia, McGill University, Montreal, QC, Canada
- Department of Anesthesia, Montreal Neurological Institute, Montreal, QC, Canada
| | - Isabelle Boileau
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Jeffrey H Meyer
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Pablo M Rusjan
- Clinical and Translational Sciences Lab, Douglas Research Centre, Montreal, QC, Canada.
- Department of Psychiatry, McGill University, Montreal, QC, Canada.
| | - Romina Mizrahi
- Clinical and Translational Sciences Lab, Douglas Research Centre, Montreal, QC, Canada.
- Department of Psychiatry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
4
|
Braga J, Kuik EJY, Lepra M, Rusjan PM, Kish SJ, Vieira EL, Nasser Z, Verhoeff N, Vasdev N, Chao T, Bagby M, Boileau I, Kloiber S, Husain MI, Kolla N, Koshimori Y, Faiz K, Wang W, Meyer JH. Astrogliosis Marker [ 11C]SL25.1188 After COVID-19 With Ongoing Depressive and Cognitive Symptoms. Biol Psychiatry 2025; 97:816-824. [PMID: 39395470 DOI: 10.1016/j.biopsych.2024.09.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/30/2024] [Accepted: 09/25/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND After acute COVID-19, 5% of people experience persistent depressive symptoms and reduced cognitive function (COVID-DC). Theoretical models propose that astrogliosis is important in long COVID, but measures primarily indicative of astrogliosis have not been studied in the brain of long COVID or COVID-DC. The objective of the current study was to measure [11C]SL25.1188 total distribution volume ([11C]SL25.1188 VT), an index of monoamine oxidase B density and a marker of astrogliosis, with positron emission tomography in participants with COVID-DC and compare with healthy control participants. METHODS In 21 COVID-DC cases and 21 healthy control participants, [11C]SL25.1188 VT was measured in the prefrontal cortex, anterior cingulate cortex, hippocampus, dorsal putamen, and ventral striatum. Depressive symptoms were measured with the Beck Depression Inventory-II, and cognitive symptoms were measured with neuropsychological tests. RESULTS [11C]SL25.1188 VT was higher in participants with COVID-DC in the prefrontal cortex, anterior cingulate cortex, hippocampus, dorsal putamen, and ventral striatum than in healthy control participants. Depressive symptom severity negatively correlated with [11C]SL25.1188 VT across prioritized brain regions. More recent acute COVID-19 positively correlated with [11C]SL25.1188 VT, reflecting higher values since predominance of the Omicron variant. Exploratory analyses found greater [11C]SL25.1188 VT in the hippocampus, dorsal putamen, and ventral striatum of COVID-DC participants than control participants with a major depressive episode with no history of COVID-19, and there was no relationship to cognitive testing in prioritized regions. CONCLUSIONS Results strongly support the presence of monoamine oxidase B-labeled astrogliosis in COVID-DC throughout the regions assessed, although the association of greater astrogliosis with fewer symptoms raises the possibility of a protective role. The magnitude of astrogliosis in COVID-DC is greater since the emergence of the Omicron variant.
Collapse
Affiliation(s)
- Joeffre Braga
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Emily J Y Kuik
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Mariel Lepra
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Pablo M Rusjan
- Douglas Research Centre and Department of Psychiatry, McGill University, Montreal, Québec, Canada
| | - Stephen J Kish
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Erica L Vieira
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Zahra Nasser
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Natasha Verhoeff
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Neil Vasdev
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Thomas Chao
- Institute of Mental Health, Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael Bagby
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Isabelle Boileau
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Stefan Kloiber
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - M Ishrat Husain
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Nathan Kolla
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Waypoint Centre for Mental Health Care, Penetanguishene, Ontario, Canada
| | - Yuko Koshimori
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Khunsa Faiz
- Department of Diagnostic Radiology, Hamilton Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Wei Wang
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Jeffrey H Meyer
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
5
|
Li MK, Yu RJ, Chen KL, Zhao Y, Yang C, Wan YJ, Long YT, Ying YL. Long-Term Real-Time Tracking of Morphology and Migration of Neuronal Cells under Oxidative Stress. CHEMICAL & BIOMEDICAL IMAGING 2025; 3:191-198. [PMID: 40151819 PMCID: PMC11938163 DOI: 10.1021/cbmi.4c00074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 03/29/2025]
Abstract
Neuronal cells exhibit diverse morphologies that are crucial for their function within the neuronal network. Long-term quantitative analysis of both neuronal cell morphology and migration is essential in neuroscience research but remains challenging. Sodium arsenite, a known inducer of oxidative stress in neurons, affects both cell morphology and migration. To rapidly assess oxidative stress in HT22 neuronal cells, we developed a method for tracking key morphological features and migration trajectories of the individual cells. Three time-dependent parameters-velocity, circularity increment, and turn angle-are identified as rapid, direct indicators of the early stages of oxidative stress in neuronal cells. This method is then applied to investigate the effects of arsenite exposure on neuronal cells. Our approach provides a valuable tool for the rapid, label-free, and long-term real-time tracking of oxidative stress in neuronal cells, offering potential insights into cellular responses under stress conditions.
Collapse
Affiliation(s)
- Ming-Kang Li
- School
of Chemistry and Chemical Engineering, Molecular Sensing and Imaging
Center (MSIC), Nanjing University, Nanjing 210023, People’s Republic of China
| | - Ru-Jia Yu
- School
of Chemistry and Chemical Engineering, Molecular Sensing and Imaging
Center (MSIC), Nanjing University, Nanjing 210023, People’s Republic of China
| | - Ke-Le Chen
- School
of Chemistry and Chemical Engineering, Molecular Sensing and Imaging
Center (MSIC), Nanjing University, Nanjing 210023, People’s Republic of China
| | - Yan Zhao
- School
of Information Science and Engineering, East China University of Science and Technology, Shanghai 200237, People’s Republic of China
| | - Cheng Yang
- School
of Electronic Sciences and Engineering, Nanjing University, Nanjing 210023, People’s
Republic of China
| | - Yong-Jing Wan
- School
of Information Science and Engineering, East China University of Science and Technology, Shanghai 200237, People’s Republic of China
| | - Yi-Tao Long
- School
of Chemistry and Chemical Engineering, Molecular Sensing and Imaging
Center (MSIC), Nanjing University, Nanjing 210023, People’s Republic of China
| | - Yi-Lun Ying
- School
of Chemistry and Chemical Engineering, Molecular Sensing and Imaging
Center (MSIC), Nanjing University, Nanjing 210023, People’s Republic of China
- Chemistry
and Biomedicine Innovation Center, Nanjing
University, Nanjing 210023, People’s
Republic of China
| |
Collapse
|
6
|
Han Y, Sun Y, Peng S, Tang T, Zhang B, Yu R, Sun X, Guo S, Ma L, Li P, Yang P. PI3K/AKT pathway: A potential therapeutic target in cerebral ischemia-reperfusion injury. Eur J Pharmacol 2025; 998:177505. [PMID: 40118329 DOI: 10.1016/j.ejphar.2025.177505] [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: 10/02/2024] [Revised: 02/21/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025]
Abstract
Cerebral ischemia is a prevalent cerebrovascular disorder, with the restoration of blocked blood vessels serving as the current standard clinical treatment. However, reperfusion can exacerbate neuronal damage and neurological dysfunction, resulting in cerebral ischemia-reperfusion (I/R) injury. Presently, clinical treatment strategies for cerebral I/R injury are limited, creating an urgent need to identify new effective therapeutic targets. The PI3K/AKT signaling pathway, a pro-survival pathway associated with cerebral I/R injury, has garnered significant attention. We conducted a comprehensive review of the literature on the PI3K/AKT pathway in the context of cerebral I/R. Our findings indicate that activation of the PI3K/AKT signaling pathway following cerebral I/R can alleviate oxidative stress, reduce endoplasmic reticulum stress (ERS), inhibit inflammatory responses, decrease neuronal apoptosis, autophagy, and pyroptosis, mitigate blood-brain barrier (BBB) damage, and promote neurological function recovery. Consequently, this pathway ultimately reduces neuronal death, alleviates brain tissue damage, decreases the volume of cerebral infarction, and improves behavioral impairments. These results suggest that the PI3K/AKT signaling pathway is a promising therapeutic target for further research and drug development, holding significant potential for the treatment of cerebral I/R injury.
Collapse
Affiliation(s)
- Yiming Han
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Yu Sun
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Shiyu Peng
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Tingting Tang
- First Clinical College, Xinxiang Medical University, Xinxiang, China
| | - Beibei Zhang
- First Clinical College, Xinxiang Medical University, Xinxiang, China
| | - Ruonan Yu
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Xiaoyan Sun
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Shanshan Guo
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China; Staff Hospital of Henan Fifth Construction Group Co., Ltd, Zhengzhou, Henan, China
| | - Lijuan Ma
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.
| | - Peng Li
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.
| | - Pengfei Yang
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.
| |
Collapse
|
7
|
García-Domínguez M. Pathological and Inflammatory Consequences of Aging. Biomolecules 2025; 15:404. [PMID: 40149940 PMCID: PMC11939965 DOI: 10.3390/biom15030404] [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: 02/25/2025] [Revised: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Aging is a complex, progressive, and irreversible biological process that entails numerous structural and functional changes in the organism. These changes affect all bodily systems, reducing their ability to respond and adapt to the environment. Chronic inflammation is one of the key factors driving the development of age-related diseases, ultimately causing a substantial decline in the functional abilities of older individuals. This persistent inflammatory state (commonly known as "inflammaging") is characterized by elevated levels of pro-inflammatory cytokines, an increase in oxidative stress, and a perturbation of immune homeostasis. Several factors, including cellular senescence, contribute to this inflammatory milieu, thereby amplifying conditions such as cardiovascular disease, neurodegeneration, and metabolic disorders. Exploring the mechanisms of chronic inflammation in aging is essential for developing targeted interventions aimed at promoting healthy aging. This review explains the strong connection between aging and chronic inflammation, highlighting potential therapeutic approaches like pharmacological treatments, dietary strategies, and lifestyle changes.
Collapse
Affiliation(s)
- Mario García-Domínguez
- Program of Immunology and Immunotherapy, CIMA-Universidad de Navarra, 31008 Pamplona, Spain;
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
8
|
Alshaebi F, Sciortino A, Kayed R. The Role of Glial Cell Senescence in Alzheimer's Disease. J Neurochem 2025; 169:e70051. [PMID: 40130281 PMCID: PMC11934031 DOI: 10.1111/jnc.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 03/26/2025]
Abstract
Glial cell senescence, characterized by the irreversible arrest of cell division and a pro-inflammatory secretory phenotype, has emerged as a critical player in the pathogenesis of Alzheimer's disease (ad). While much attention has been devoted to the role of neurons in ad, growing evidence suggests that glial cells, including astrocytes, microglia, and oligodendrocytes, contribute significantly to disease progression through senescence. In this review, we explore the molecular mechanisms underlying glial cell senescence in ad, focusing on the cellular signaling pathways, including DNA damage response and the accumulation of senescence-associated secretory phenotypes (SASP). We also examine how senescent glial cells exacerbate neuroinflammation, disrupt synaptic function, and promote neuronal death in ad. Moreover, we discuss emerging therapeutic strategies aimed at targeting glial cell senescence to mitigate the neurodegenerative processes in ad. By providing a comprehensive overview of current research on glial cell senescence in Alzheimer's disease, this review highlights its potential as a novel therapeutic target in the fight against ad.
Collapse
Affiliation(s)
- Fadhl Alshaebi
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Alessia Sciortino
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
| |
Collapse
|
9
|
Chen W, Mao T, Ma R, Xiong Y, Han R, Wang L. The role of astrocyte metabolic reprogramming in ischemic stroke (Review). Int J Mol Med 2025; 55:49. [PMID: 39930815 PMCID: PMC11781528 DOI: 10.3892/ijmm.2025.5490] [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: 10/24/2024] [Accepted: 01/08/2025] [Indexed: 02/13/2025] Open
Abstract
Ischemic stroke, a leading cause of disability and mortality worldwide, is characterized by the sudden loss of blood flow in specific area of the brain. Intravenous thrombolysis with recombinant tissue plasminogen activator is the only approved pharmacological treatment for acute ischemic stroke; however, the aforementioned treatment has significant clinical limitations, thus there is an urgent need for the development of novel mechanisms and therapeutic strategies for ischemic stroke. Astrocytes, abundant and versatile cells in the central nervous system, offer crucial support to neurons nutritionally, structurally and physically. They also contribute to blood‑brain barrier formation and regulate neuronal extracellular ion concentrations. Accumulated evidence has revealed the involvement of astrocytes in the regulation of host neurotransmitter metabolism, immune response and tissue repair, and different metabolic characteristics of astrocytes can contribute to the process and development of ischemic stroke, suggesting that targeted regulation of astrocyte metabolic reprogramming may contribute to the treatment and prognosis of ischemic stroke. In the present review, the current understanding of the multifaceted mechanisms of astrocyte metabolic reprogramming in ischemic stroke, along with its regulatory factors and pathways, as well as the strategies to promote its polarization balance, which hold promise for astrocyte immunometabolism‑targeted therapies in the treatment of ischemic stroke, were summarized.
Collapse
Affiliation(s)
- Weixin Chen
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100105, P.R. China
| | - Tangyou Mao
- Gastroenterology Department, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, P.R. China
| | - Rui Ma
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100105, P.R. China
| | - Yuxuan Xiong
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100105, P.R. China
| | - Ran Han
- Clinical Laboratory Department, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, P.R. China
| | - Le Wang
- Cerebrovascular Disease Department, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, P.R. China
| |
Collapse
|
10
|
Mazowiecki M, Flet-Berliac L, Roux J, Lépine A, Chretien P, Hacein-Bey-Abina S, Giorgi L, Villega F, Cheuret E, Benaiteau M, Rogemond V, Picard G, Baer S, Cleuziou P, Lametery E, Desguerre I, Aubart M, Chevignard M, Le Grand R, Horellou P, Leroy C, Joubert B, Honnorat J, Deiva K. Long-Term Clinical and Biological Prognostic Factors of Anti-NMDA Receptor Encephalitis in Children. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2025; 12:e200346. [PMID: 39715492 DOI: 10.1212/nxi.0000000000200346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 11/22/2024] [Indexed: 12/25/2024]
Abstract
BACKGROUND AND OBJECTIVES Anti-NMDAR encephalitis (NMDARE) is a severe neurologic condition, and recently, the NMDAR Encephalitis One-Year Functional Status (NEOS) score has emerged as a 1-year prognostic tool. This study aimed to evaluate NEOS score and biomarker (neurofilament light chains [NfL], total-Tau protein, glial fibrillary acidic protein, and serum cytokines) correlation with modified Rankin Scale (mRS), cognitive impairment, and clinical recovery in pediatric NMDARE over 2 years. METHODS In this French multicenter observational study, 104 pediatric patients with NMDARE were followed for a minimum of 2 years. Clinical data and serum/plasma samples were collected. Biomarker levels, measured using electroluminescence mesoscale discovery (MSD) S-PLEX, were compared between patients and controls and assessed for correlations with disease activity, mRS, cognitive/language impairment, and recovery status at 2 years. RESULTS At a median follow-up of 39.5 months, 68 percent of patients had unfavorable recovery and 54% had significant cognitive impairment. Both outcomes were strongly associated with younger age at diagnosis (OR 6.10 [1.91-27.3] p < 0.01 and 5.69 [1.46-27.7] p = 0.02, respectively). A higher NEOS score was significantly correlated with increased cognitive impairment (OR 2.53 [1.52-4.21], p < 0.001), higher mRS scores (OR 2.12 [1.34-3.57], p < 0.01), and unfavorable recovery at 2 years (OR 2.00 [1.30-3.06], p = 0.015). Elevated NfL levels were significantly associated with unfavorable recovery (OR 3.62 [1.29-10.9] p = 0.012) and severe cognitive impairment (OR 3.77 [1.38-10.9] p = 0.012) at 2 years. The combined area under the curve (AUC) for NfL and NEOS was significantly higher than the AUCs of NEOS and NfL alone (p = 0.01). DISCUSSION The NEOS score strongly predicts long-term outcomes in NMDARE, with its predictive value extending beyond the first-year mR prediction. NfL levels at disease onset seem to improve accuracy in predicting poor outcomes, providing valuable information for treatment decisions and future clinical trials.
Collapse
Affiliation(s)
- Maxime Mazowiecki
- Pediatric Neurology Departement, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University Hospitals, Bicêtre Hospital, and Paris-Saclay University, Le Kremlin-Bicêtre
| | - Lorraine Flet-Berliac
- Pediatric Neurology Departement, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University Hospitals, Bicêtre Hospital, and Paris-Saclay University, Le Kremlin-Bicêtre
| | - Julia Roux
- Pediatric Neurology Departement, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University Hospitals, Bicêtre Hospital, and Paris-Saclay University, Le Kremlin-Bicêtre
| | - Anne Lépine
- Pediatric Neurology Department, Assistance Publique des Hôpitaux de Marseille, Hôpital Universitaire, Marseille
| | - Pascale Chretien
- Clinical Immunology Laboratory, Assistance Publique des Hôpitaux de Paris, Hôpitaux Universitaires Paris-Saclay, Bicêtre Hospital, and Paris-Saclay University, Le Kremlin-Bicêtre
- UTCBS, UMR8258 CNRS-U1267 INSERM, Faculté de Pharmacie de Paris, Université de Paris
| | - Salima Hacein-Bey-Abina
- Clinical Immunology Laboratory, Assistance Publique des Hôpitaux de Paris, Hôpitaux Universitaires Paris-Saclay, Bicêtre Hospital, and Paris-Saclay University, Le Kremlin-Bicêtre
- UTCBS, UMR8258 CNRS-U1267 INSERM, Faculté de Pharmacie de Paris, Université de Paris
| | - Laetitia Giorgi
- Pediatric Neurology Departement, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University Hospitals, Bicêtre Hospital, and Paris-Saclay University, Le Kremlin-Bicêtre
- National Referral Center for Rare Inflammatory Brain and Spinal Diseases, Le Kremlin-Bicêtre; and
| | - Frederic Villega
- Pediatric Neurology Department, CICp-1401, University Children Hospital, Bordeaux
- Interdisciplinary Institute for Neurosciences, CNRS UMR 5297
| | - Emmanuel Cheuret
- Pediatric Neurology Department, Purpan University Hospital, Toulouse
| | - Marie Benaiteau
- Reference Center on autoimmune encephalitis, Hospices Civils de Lyon, Institut MELIS, Inserm U1314/CNRS UMR 5284, Université Claude Bernard Lyon 1
| | - Veronique Rogemond
- Reference Center on autoimmune encephalitis, Hospices Civils de Lyon, Institut MELIS, Inserm U1314/CNRS UMR 5284, Université Claude Bernard Lyon 1
| | - Geraldine Picard
- Reference Center on autoimmune encephalitis, Hospices Civils de Lyon, Institut MELIS, Inserm U1314/CNRS UMR 5284, Université Claude Bernard Lyon 1
| | - Sarah Baer
- Department of Neuropediatrics, ERN EpiCare, Hôpitaux Universitaires de Strasbourg
- Institute for Genetics and Molecular and Cellular Biology (IGBMC), University of Strasbourg, CNRS UMR7104, INSERM U1258, Illkirch, France
| | - Pierre Cleuziou
- Department of Pediatric Neurology, Lille University Hospital
| | - Elodie Lametery
- Pediatric Department, Grenoble Alpes University Hospital, Hôpital Albert Michallon
| | - Isabelle Desguerre
- Pediatric Neurology Department Necker-Enfants Malades Hospital, University of Paris, AP-HP
| | - Mélodie Aubart
- Pediatric Neurology Department Necker-Enfants Malades Hospital, University of Paris, AP-HP
| | - Mathilde Chevignard
- Rehabilitation Department for Children with Acquired Neurological Injury, Saint-Maurice Hospitals (M.C.); Saint Maurice Hospitals
- Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale (LIB)
- Sorbonne Université, GRC 24 Handicap Moteur Cognitif et Réadaptation (HaMCRe), Paris
| | - Roger Le Grand
- Université Paris-Saclay, CEA, INSERM Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT)
| | - Philippe Horellou
- Université Paris-Saclay, CEA, INSERM Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT)
- National Referral Center for Rare Inflammatory Brain and Spinal Diseases, Le Kremlin-Bicêtre; and
| | - Carole Leroy
- Université Paris-Saclay, CEA, INSERM Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT)
- National Referral Center for Rare Inflammatory Brain and Spinal Diseases, Le Kremlin-Bicêtre; and
| | - Bastien Joubert
- Reference Center on autoimmune encephalitis, Hospices Civils de Lyon, Institut MELIS, Inserm U1314/CNRS UMR 5284, Université Claude Bernard Lyon 1
| | - Jerome Honnorat
- Reference Center on autoimmune encephalitis, Hospices Civils de Lyon, Institut MELIS, Inserm U1314/CNRS UMR 5284, Université Claude Bernard Lyon 1
| | - Kumaran Deiva
- Pediatric Neurology Departement, Assistance Publique-Hôpitaux de Paris, Paris-Saclay University Hospitals, Bicêtre Hospital, and Paris-Saclay University, Le Kremlin-Bicêtre
- Université Paris-Saclay, CEA, INSERM Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT)
- National Referral Center for Rare Inflammatory Brain and Spinal Diseases, Le Kremlin-Bicêtre; and
- Institut Universitaire de France, France
| |
Collapse
|
11
|
Soares JRP, dos Santos CC, de Oliveira LMG, Rocha Neto H, Victor MM, França EL, Costa MDFD, Costa SL, de Oliveira JVR. Synthesis of Naringenin and Senecioic Acid Ester Derivatives and Biological Evaluation of the Astrocyte Antioxidant Mechanism and Reactivity After Inflammatory Stimulus. Int J Mol Sci 2025; 26:2215. [PMID: 40076834 PMCID: PMC11900193 DOI: 10.3390/ijms26052215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
The imbalance between the overproduction of reactive species and antioxidant mechanisms can result in astrogliosis and oxidative stress associated with neurodegeneration. Based on the described antioxidant activity of naturally occurring flavonoids, this study evaluated the antioxidant mechanisms of the flavonoid naringenin and the senecioic acid ester derivatives in cortical astrocytes. Naringenin and (S)-naringenin were purified from Citrus paradisi, and from them 7,4-O-disenecioic ester naringenin, (S)-7,4-O-disenecioic ester naringenin, and 7-O-senecioic ester naringenin were synthesized and tested for antioxidant activity by the free-radical scavenging reaction with DPPH. The flavonoids' toxicity and glutathione (GS) depletion were determined in rat astrocyte cultures; the effects on the astrocytes' reactivity was determined by the expression of the glial fibrillary acidic protein (GFAP) and by measuring nitric oxide (NO) production in astrocytes treated with lipopolysaccharide (LPS, 1 µg/mL/24 h). The compounds (1-10 μM) presented antioxidant effects, and the (S)-7,4'-O-disenecioic ester naringenin was the most effective. The compounds (1-100 μM) were not toxic to the astrocytes, also promoting an antioxidant effect by increasing GSH. Moreover, naringenin, (S)-7,4'-O-disenecioic ester naringenin, and 7-O-senecioc ester naringenin mitigated the astrocyte reactivity induced by LPS, reducing GFAP expression and NO production. These findings indicate that naringenin and senecioic acid ester derivatives present a pharmacological potential as antioxidant and anti-inflammatory compounds for brain diseases via the modulation of astrocyte response.
Collapse
Affiliation(s)
- Janaína Ribeiro Pereira Soares
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Brazil; (J.R.P.S.); (C.C.d.S.); (L.M.G.d.O.); (H.R.N.); (M.d.F.D.C.)
| | - Cleonice Creusa dos Santos
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Brazil; (J.R.P.S.); (C.C.d.S.); (L.M.G.d.O.); (H.R.N.); (M.d.F.D.C.)
| | - Lucas Matheus Gonçalves de Oliveira
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Brazil; (J.R.P.S.); (C.C.d.S.); (L.M.G.d.O.); (H.R.N.); (M.d.F.D.C.)
| | - Heráclito Rocha Neto
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Brazil; (J.R.P.S.); (C.C.d.S.); (L.M.G.d.O.); (H.R.N.); (M.d.F.D.C.)
| | - Maurício Moraes Victor
- Department of Organic Chemistry, Institute of Chemistry, Federal University of Bahia, Salvador 40170-115, Brazil;
| | - Elivana Lima França
- Federal Institute of Bahia, Campus Vitória da Conquista, Vitória da Conquista 45078-300, Brazil;
| | - Maria de Fátima Dias Costa
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Brazil; (J.R.P.S.); (C.C.d.S.); (L.M.G.d.O.); (H.R.N.); (M.d.F.D.C.)
- National Institute of Translational Neuroscience (INNT), Rio de Janeiro 21941-902, Brazil
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Brazil; (J.R.P.S.); (C.C.d.S.); (L.M.G.d.O.); (H.R.N.); (M.d.F.D.C.)
- National Institute of Translational Neuroscience (INNT), Rio de Janeiro 21941-902, Brazil
| | - Juciele Valeria Ribeiro de Oliveira
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Brazil; (J.R.P.S.); (C.C.d.S.); (L.M.G.d.O.); (H.R.N.); (M.d.F.D.C.)
| |
Collapse
|
12
|
Abbott V, Housden BE, Houldsworth A. Could immunotherapy and regulatory T cells be used therapeutically to slow the progression of Alzheimer's disease? Brain Commun 2025; 7:fcaf092. [PMID: 40078868 PMCID: PMC11896979 DOI: 10.1093/braincomms/fcaf092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/25/2024] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Alzheimer's disease and other cognitive impairments are a growing problem in the healthcare world with the ageing population. There are currently no effective treatments available; however, it has been suggested that targeting neuroinflammation may be a successful approach in slowing the progression of neurodegeneration. Reducing the destructive hyperinflammatory pathology to maintain homeostasis in neural tissue is a promising option to consider. This review explores the mechanisms behind neuroinflammation and the effectiveness of immunotherapy in slowing the progression of cognitive decline in patients with Alzheimer's disease. The key components of neuroinflammation in Alzheimer's disease researched are microglia, astrocytes, cytokines and CD8+ effector T cells. The role of oxidative stress on modulating regulatory T cells and some of the limitations of regulatory T cell-based therapies are also explored. Increasing regulatory T cells can decrease activation of microglia, proinflammatory cytokines and astrocytes; however, it can also increase levels of inflammatory cytokines. There is a complex network of regulatory T cell interactions that reduce Alzheimer's disease pathology, which is not fully understood. Exploring the current literature, further research into the use of immunotherapy in Alzheimer's disease is vital to determine the potential of these techniques; however, there is sufficient evidence to suggest that increasing regulatory T cells count does prevent Alzheimer's disease symptoms and pathology in patients with Alzheimer's disease. Some exciting innovative therapies are muted to explore in the future. The function of regulatory T cells in the presence of reactive oxygen species and oxidative stress should be investigated further in patients with neurogenerative disorders to ascertain if combination therapies could reduce oxidative stress while also enhancing regulatory T cells function. Could methods of immunotherapy infuse exogenous functional Tregs or enhance the immune environment in favour of endogenous regulatory T cells differentiation, thus reducing neuroinflammation in neurodegenerative pathology, inhibiting the progression of Alzheimer's disease?
Collapse
Affiliation(s)
- Victoria Abbott
- Neuroscience, Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter EX2 4TH, UK
| | - Benjamin E Housden
- Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter EX2 4TH, UK
- Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK
| | - Annwyne Houldsworth
- Neuroscience, Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter EX2 4TH, UK
- Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter EX2 4TH, UK
| |
Collapse
|
13
|
Gallagher E, Li S, Lee H, Xu H, Lee VMY, Mach RH, McManus MJ. Noninvasive Detection of Oxidative Stress in a Mouse Model of 4R Tauopathy via Positron Emission Tomography with [ 18F]ROStrace. Int J Mol Sci 2025; 26:1845. [PMID: 40076472 PMCID: PMC11899037 DOI: 10.3390/ijms26051845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Oxidative stress, defined as the excessive production of reactive oxygen species (ROS), is a crucial factor in the pathogenesis of various neurodegenerative diseases, including the 4-repeat (4R) tauopathies. Collectively, the 4R tauopathies are characterized by the progressive aggregation of tau protein isoforms with four microtubule-binding domains in and around brain cells. The cyclical relationship between oxidative stress and 4R tau aggregation suggests that a means of imaging ROS noninvasively could be a valuable tool for the study and treatment of 4R tauopathy in both humans and animal models. To demonstrate the potential of the ROS-sensitive positron emission tomography (PET) radiotracer [18F]ROStrace as a means of filling this methodological gap, we performed [18F]ROStrace PET imaging on PS19 mice, which exhibit 4R tau aggregation similar to that seen in human 4R tauopathy. Significant increases in [18F]ROStrace signal became detectable in the hippocampus of 6-11-month-old (mo) PS19 animals and spread to the brainstem, midbrain, and thalamus of 11+ mo animals. Additionally, older PS19 mice displayed higher whole-brain average [18F]ROStrace signal compared to age-matched controls (p = 0.042), and tau pathology consistently colocalized with multiple fluorescent indicators of oxidative stress in PS19 brain samples. These results provide novel evidence that 4R tau aggregation is associated with increased oxidative stress in PS19 mouse brain and advance [18F]ROStrace as a noninvasive technology for the detection of oxidative stress in neurodegenerative diseases involving tau pathology.
Collapse
Affiliation(s)
- Evan Gallagher
- Department of Anesthesia and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA (R.H.M.)
| | - Shihong Li
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA (R.H.M.)
| | - Hsiaoju Lee
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA (R.H.M.)
| | - Hong Xu
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Virginia M.-Y. Lee
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert H. Mach
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA (R.H.M.)
| | - Meagan J. McManus
- Department of Anesthesia and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| |
Collapse
|
14
|
Hoeferlin GF, Grabinski SE, Druschel LN, Duncan JL, Burkhart G, Weagraff GR, Lee AH, Hong C, Bambroo M, Olivares H, Bajwa T, Coleman J, Li L, Memberg W, Sweet J, Hamedani HA, Acharya AP, Hernandez-Reynoso AG, Donskey C, Jaskiw G, Ricky Chan E, Shoffstall AJ, Bolu Ajiboye A, von Recum HA, Zhang L, Capadona JR. Bacteria invade the brain following intracortical microelectrode implantation, inducing gut-brain axis disruption and contributing to reduced microelectrode performance. Nat Commun 2025; 16:1829. [PMID: 39979293 PMCID: PMC11842729 DOI: 10.1038/s41467-025-56979-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 01/29/2025] [Indexed: 02/22/2025] Open
Abstract
Brain-machine interface performance can be affected by neuroinflammatory responses due to blood-brain barrier (BBB) damage following intracortical microelectrode implantation. Recent findings suggest that certain gut bacterial constituents might enter the brain through damaged BBB. Therefore, we hypothesized that damage to the BBB caused by microelectrode implantation could facilitate microbiome entry into the brain. In our study, we found bacterial sequences, including gut-related ones, in the brains of mice with implanted microelectrodes. These sequences changed over time. Mice treated with antibiotics showed a reduced presence of these bacteria and had a different inflammatory response, which temporarily improved microelectrode recording performance. However, long-term antibiotic use worsened performance and disrupted neurodegenerative pathways. Many bacterial sequences found were not present in the gut or in unimplanted brains. Together, the current study established a paradigm-shifting mechanism that may contribute to chronic intracortical microelectrode recording performance and affect overall brain health following intracortical microelectrode implantation.
Collapse
Grants
- R01 NS131502 NINDS NIH HHS
- R25 CA221718 NCI NIH HHS
- T32 EB004314 NIBIB NIH HHS
- This study was supported in part by Merit Review Award GRANT12418820 (Capadona), Biomedical Science and Engineering Summer Program for Rehabilitation Interventions GRANT14089804 (Capadona/Hess-Dunning), and Senior Research Career Scientist Award # GRANT12635707 (Capadona) from the United States (US) Department of Veterans Affairs Rehabilitation Research and Development Service. Additionally, this work was also supported in part by the National Institute of Health, National Institute of Neurological Disorders and Stroke GRANT12635723 (Capadona/Pancrazio and diversity supplement Hernandez-Reynoso) and NS131502 (Ware/Pancrazio/Capadona), the National Cancer Institute NCI R25 CA221718 (Berger) provided support for Weagraff, the Congressionally Directed Medical Research Program (CDMRP) – Spinal Cord Injury Research Program (SCIRP), administered through the Department of Defense Award # SC180308 (Ajiboye) and the National Institute for Biomedical Imaging and Bioengineering, T32EB004314, provided support for both Hoeferlin and Burkhart (Capadona/Kirsch). Microbiome analyses were partially supported by the junior faculty’s startup funding from the CWRU School of Medicine, BGT630267 (Zhang). Finally, partial funding was provided from discretionary funding from the Donnell Institute Professorship endowment (Capadona) and the Case School of Engineering Research Incentive Program (Capadona).
Collapse
Affiliation(s)
- George F Hoeferlin
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Sarah E Grabinski
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Lindsey N Druschel
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Jonathan L Duncan
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Grace Burkhart
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Gwendolyn R Weagraff
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
- Department of Biology, University of Florida, Gainesville, FL, USA
| | - Alice H Lee
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Christopher Hong
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Meera Bambroo
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Hannah Olivares
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Tejas Bajwa
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Jennifer Coleman
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Longshun Li
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - William Memberg
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Jennifer Sweet
- Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
- Department of Neurological Surgery, University Hospitals Case Medical Center, Cleveland, OH, USA
| | - Hoda Amani Hamedani
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
- Department of Materials Science and Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Abhinav P Acharya
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Ana G Hernandez-Reynoso
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, USA
| | - Curtis Donskey
- Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
- Division of Infectious Diseases & HIV Medicine in the Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - George Jaskiw
- Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA
| | - E Ricky Chan
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Andrew J Shoffstall
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - A Bolu Ajiboye
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Horst A von Recum
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Liangliang Zhang
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA.
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA.
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA.
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA.
| |
Collapse
|
15
|
Mesentier-Louro LA, Goldman C, Ndayisaba A, Buonfiglioli A, Rooklin RB, Schuldt BR, Uchitelev A, Khurana V, Blanchard JW. Cholesterol-mediated Lysosomal Dysfunction in APOE4 Astrocytes Promotes α-Synuclein Pathology in Human Brain Tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.09.637107. [PMID: 39975381 PMCID: PMC11839026 DOI: 10.1101/2025.02.09.637107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The pathological hallmark of neurodegenerative disease is the aberrant post-translational modification and aggregation of proteins leading to the formation of insoluble protein inclusions. Genetic factors like APOE4 are known to increase the prevalence and severity of tau, amyloid, and α-Synuclein inclusions. However, the human brain is largely inaccessible during this process, limiting our mechanistic understanding. Here, we developed an iPSC-based 3D model that integrates neurons, glia, myelin, and cerebrovascular cells into a functional human brain tissue (miBrain). Like the human brain, we found pathogenic phosphorylation and aggregation of α-Synuclein is increased in the APOE4 miBrain. Combinatorial experiments revealed that lipid-droplet formation in APOE4 astrocytes impairs the degradation of α-synuclein and leads to a pathogenic transformation that seeds neuronal inclusions of α-Synuclein. Collectively, this study establishes a robust model for investigating protein inclusions in human brain tissue and highlights the role of astrocytes and cholesterol in APOE4-mediated pathologies, opening therapeutic opportunities.
Collapse
Affiliation(s)
- Louise A. Mesentier-Louro
- Icahn School of Medicine, Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- These authors contributed equally
| | - Camille Goldman
- Icahn School of Medicine, Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- These authors contributed equally
| | - Alain Ndayisaba
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Movement Disorders, American Parkinson Disease Association (APDA) Center for Advanced Research and MSA Center of Excellence, Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Alice Buonfiglioli
- Icahn School of Medicine, Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY, USA
| | - Rikki B. Rooklin
- Icahn School of Medicine, Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY, USA
| | - Braxton R. Schuldt
- Icahn School of Medicine, Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY, USA
| | - Abigail Uchitelev
- Icahn School of Medicine, Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY, USA
- Macaulay Honors College at Hunter College, New York, NY, USA
| | - Vikram Khurana
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Movement Disorders, American Parkinson Disease Association (APDA) Center for Advanced Research and MSA Center of Excellence, Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Joel W. Blanchard
- Icahn School of Medicine, Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Lead contact
| |
Collapse
|
16
|
Carrasco M, Guzman L, Olloquequi J, Cano A, Fortuna A, Vazquez-Carrera M, Verdaguer E, Auladell C, Ettcheto M, Camins A. Licochalcone A prevents cognitive decline in a lipopolysaccharide-induced neuroinflammation mice model. Mol Med 2025; 31:54. [PMID: 39930360 PMCID: PMC11812219 DOI: 10.1186/s10020-025-01106-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/27/2025] [Indexed: 02/13/2025] Open
Abstract
Inflammation plays a key role in the development of neurodegenerative disorders that are currently incurable. Licochalcone A (LCA) has been described as an emerging anti-inflammatory drug with multiple therapeutical properties that could potentially prevent neurodegeneration. However, its neuroprotective mechanism remains unclear. Here, we investigated if LCA prevents cognitive decline induced by Lipopolysaccharide (LPS) and elucidated its potential benefits. For that, 8-week-old C57BL6/J male mice were intraperitonially (i.p.) treated with saline solution or LCA (15 mg/kg/day, 3 times per week) for two weeks. The last day, a single i.p injection of LPS (1 mg/kg) or saline solution was administered 24 h before sacrifice. The results revealed a significant reduction in mRNA expression in genes involved in oxidative stress (Sod1, Cat, Pkm, Pdha1, Ndyfv1, Uqcrb1, Cycs and Cox4i1), metabolism (Slc2a1, Slc2a2, Prkaa1 and Gsk3b) and synapsis (Bdnf, Nrxn3 and Nlgn2) in LPS group compared to saline. These findings were linked to memory impairment and depressive-like behavior observed in this group. Interestingly, LCA protected against LPS alterations through its anti-inflammatory effect, reducing gliosis and regulating M1/M2 markers. Moreover, LCA-treated animals showed a significant improvement of antioxidant mechanisms, such as citrate synthase activity and SOD2. Additionally, LCA demonstrated protection against metabolic disturbances, downregulating GLUT4 and P-AKT, and enhanced the expression of synaptic-related proteins (P-CREB, BDNF, PSD95, DBN1 and NLG3), leading all together to dendritic spine preservation. In conclusion, our results demonstrate that LCA treatment prevents LPS-induced cognitive decline by reducing inflammation, enhancing the antioxidant response, protecting against metabolic disruptions and improving synapsis related mechanisms.
Collapse
Affiliation(s)
- Marina Carrasco
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Laura Guzman
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
| | - Jordi Olloquequi
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Talca, Chile
| | - Amanda Cano
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Ana Fortuna
- Laboratory of Pharmacology and Pharmaceutical Care, Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, CIBIT/ICNAS, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Manuel Vazquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain
- Networking Research Centre of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, 28031, Madrid, Spain
- Institute of Biomedicine of the Universitat de Barcelona (IBUB), University of Barcelona, 08028, Barcelona, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, 08950, Esplugues de Llobregat, Spain
| | - Ester Verdaguer
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Carme Auladell
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain.
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain.
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain.
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028, Barcelona, Spain
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| |
Collapse
|
17
|
Samà M, Musillo C, Cirulli F. Counteracting the effects of maternal obesity on offspring neurodevelopment through Omega-3-based nutritional strategies. Neuroscience 2025; 566:142-148. [PMID: 39722288 DOI: 10.1016/j.neuroscience.2024.12.041] [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: 10/01/2024] [Revised: 12/09/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024]
Abstract
It is becoming increasingly recognized that, in addition to psychological stress, unbalanced maternal nutritional habits can threaten fetal brain development. Maternal obesity is one of the most pressing public health problems facing the world today, as about 40% of pregnant women are obese or gain excessive weight worldwide. This condition can negatively impact offspring's brain development, increasing the risk for autism spectrum disorders, cognitive deficits, attention deficit hyperactivity disorder, as well as anxiety and depression. In the context of fetal development, nutritional interventions may represent a feasible and safe approach for preventing the negative effects of maternal obesity. We argue that maternal Omega-3 supplementation, among the many dietary strategies available, is especially promising as it buffers oxidative stress and inflammation, both recognized as candidate mechanisms underlying the negative long-term effects of maternal obesity on the offspring. Notwithstanding the current knowledge, both preclinical studies and clinical trials are needed to refine current strategies addressing dietary content and length of administration according to individual characteristics and needs.
Collapse
Affiliation(s)
- Marianna Samà
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299 00161, Rome, Italy
| | - Chiara Musillo
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299 00161, Rome, Italy.
| | - Francesca Cirulli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299 00161, Rome, Italy
| |
Collapse
|
18
|
Thergarajan P, O'Brien TJ, Jones NC, Ali I. Ligand-receptor interactions: A key to understanding microglia and astrocyte roles in epilepsy. Epilepsy Behav 2025; 163:110219. [PMID: 39693861 DOI: 10.1016/j.yebeh.2024.110219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/30/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024]
Abstract
Epilepsy continues to pose significant social and economic challenges on a global scale. Existing therapeutic approaches predominantly revolve around neurocentric mechanisms, and fail to control seizures in approximately one-third of patients. This underscores the pressing need for novel and complementary treatment approaches to address this gap. An increasing body of literature points to a role for glial cells, including microglia and astrocytes, in the pathogenesis of epilepsy. Notably, microglial cells, which serve as pivotal inflammatory mediators within the epileptic brain, have received increasing attention over recent years. These immune cells react to epileptogenic insults, regulate neuronal processes, and play diverse roles during the process of epilepsy development. Additionally, astrocytes, another integral non-neuronal brain cells, have garnered increasing recognition for their dynamic contributions to the pathophysiology of epilepsy. Their complex interactions with neurons and other glial cells involve modulating synaptic activity and neuronal excitability, thereby influencing the aberrant networks formed during epileptogenesis. This review explores the alterations in microglial and astrocytic function and their mechanisms of communication following an epileptogenic insult, examining their contribution to epilepsy development. By comprehensively studying these mechanisms, potential avenues could emerge for refining therapeutic strategies and ameliorating the impact of this complex neurological disease.
Collapse
Affiliation(s)
- Peravina Thergarajan
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, 3004, Australia
| | - Terence J O'Brien
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, 3004, Australia; Department of Neurology, The Alfred Hospital, Melbourne, Victoria, 3004, Australia; Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Victoria, 3000, Australia
| | - Nigel C Jones
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, 3004, Australia; Department of Neurology, The Alfred Hospital, Melbourne, Victoria, 3004, Australia; Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Victoria, 3000, Australia
| | - Idrish Ali
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, 3004, Australia; Department of Neurology, The Alfred Hospital, Melbourne, Victoria, 3004, Australia; Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Victoria, 3000, Australia
| |
Collapse
|
19
|
Liu Q, Tan Y, Zhang ZW, Tang W, Han L, Peng KP, Liu MH, Tian GX. The role of NLRP3 inflammasome-mediated pyroptosis in astrocytes during hyperoxia-induced brain injury. Inflamm Res 2025; 74:25. [PMID: 39862240 DOI: 10.1007/s00011-024-01984-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/08/2024] [Accepted: 10/18/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Hyperoxia-induced brain injury is a severe neurological complication that is often accompanied by adverse long-term prognosis. The pathogenesis of hyperoxia-induced brain injury is highly complex, with neuroinflammation playing a crucial role. The activation of the nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome, which plays a pivotal role in regulating and amplifying the inflammatory response, is the pathological core of hyperoxia-induced brain injury. Additionally, astrocytes actively participate in neuroinflammatory responses. However, there is currently no comprehensive overview summarizing the role of astrocytes in hyperoxia-induced brain injury and the NLRP3 signaling pathways in astrocytes. OBJECTIVE This article aims to provide an overview of studies reported in the literature investigating the pathological role of astrocyte involvement during the inflammatory response in hyperoxia-induced brain injury, the mechanisms of hyperoxia activateing the NLRP3 inflammasome to mediate pyroptosis in astrocytes, and the potential therapeutic effects of drugs targeting the NLRP3 inflammasome to alleviate hyperoxia-induced brain injury. METHOD We searched major databases (including PubMed, Web of Science, and Google Scholar, etc.) for literature encompassing astrocytes, NLRP3 inflammasome, and pyroptosis during hyperoxia-induced brain injury up to Oct 2024. We combined with studies found in the reference lists of the included studies. CONCLUSION In this study, we elucidated the transition of function in astrocytes and activation mechanisms under hyperoxic conditions, and we summarized the potential upstream of the trigger involved in NLRP3 inflammasome activation during hyperoxia-induced brain injury, such as ROS and potassium efflux. Furthermore, we described the signaling pathways of the NLRP3 inflammasome and pyroptosis executed by GSDMD and GSDME in astrocytes under hyperoxic conditions. Finally, we summarized the inhibitors targeting the NLRP3 inflammasome in astrocytes to provide new insights for treating hyperoxia-induced brain injury.
Collapse
Affiliation(s)
- Qiao Liu
- Department of Ultrasound, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Yan Tan
- Department of Ultrasound, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Zhan-Wei Zhang
- Department of Neurosurgery, The First Hospital of Hunan University of Chinese Medicine, 40007, Changsha, China
| | - Wang Tang
- Department of Ultrasound, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Lei Han
- Department of Ultrasound, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Ke-Ping Peng
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Hospital of Hunan University of Chinese Medicine, 40007, Changsha, China
| | - Ming-Hui Liu
- Department of Ultrasound, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Gui-Xiang Tian
- Department of Ultrasound, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
| |
Collapse
|
20
|
Padti AC, Bhavi SM, Thokchom B, Singh SR, Bhat SS, Harini BP, Sillanpää M, Yarajarla RB. Nanoparticle Interactions with the Blood Brain Barrier: Insights from Drosophila and Implications for Human Astrocyte Targeted Therapies. Neurochem Res 2025; 50:80. [PMID: 39832031 DOI: 10.1007/s11064-025-04333-x] [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: 11/09/2024] [Revised: 12/11/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
This review explores the intricate connections between Drosophila models and the human blood-brain barrier (BBB) with nanoparticle-based approaches for neurological treatment. Drosophila serves as a powerful model organism due to its evolutionary conservation of key biological processes, particularly in the context of the BBB, which is formed by glial cells that share structural and functional similarities with mammalian endothelial cells. Recent advancements in nanoparticle technology have highlighted their potential for effective drug delivery across the BBB, utilizing mechanisms such as passive diffusion, receptor-mediated transcytosis, and carrier-mediated transport. The ability to engineer nanoparticles with specific physicochemical properties-such as size, surface charge, and functionalization-enhances their targeting capabilities, particularly towards astrocytes, which play a crucial role in maintaining BBB integrity and responding to neuroinflammation. Insights gained from Drosophila studies have informed the design of personalized nanomedicine strategies aimed at treating neurodegenerative diseases, including Alzheimer's, Parkinson's disease etc. As research progresses, the integration of findings from Drosophila models with emerging humanized BBB systems will pave the way for innovative therapeutic approaches that improve drug delivery and patient outcomes in neurological disorders.
Collapse
Affiliation(s)
- Akshata Choudhari Padti
- Drosophila and Nanoscience Research Laboratory, Department of Applied Genetics, Karnatak University, Dharwad, Karnataka, 580003, India
| | - Santosh Mallikarjun Bhavi
- Drosophila and Nanoscience Research Laboratory, Department of Applied Genetics, Karnatak University, Dharwad, Karnataka, 580003, India
| | - Bothe Thokchom
- Drosophila and Nanoscience Research Laboratory, Department of Applied Genetics, Karnatak University, Dharwad, Karnataka, 580003, India
| | - Sapam Riches Singh
- Drosophila and Nanoscience Research Laboratory, Department of Applied Genetics, Karnatak University, Dharwad, Karnataka, 580003, India
| | - Shivanand S Bhat
- Department of Botany, Smt. Indira Gandhi Government First Grade Women's College, Sagar, Karnataka, 577401, India
| | - B P Harini
- Department of Zoology and Centre for Applied Genetics, Bangalore University, Bangaluru, Karnataka, 560056, India
| | - Mika Sillanpää
- Department of Biological and Chemical Engineering, Aarhus University, Norrebrogade 44, Aarhus C, 8000, Denmark
| | - Ramesh Babu Yarajarla
- Drosophila and Nanoscience Research Laboratory, Department of Applied Genetics, Karnatak University, Dharwad, Karnataka, 580003, India.
| |
Collapse
|
21
|
Bhalla M, Joo J, Kim D, Shin JI, Park YM, Ju YH, Park U, Yoo S, Hyeon SJ, Lee H, Lee J, Ryu H, Lee CJ. SIRT2 and ALDH1A1 as critical enzymes for astrocytic GABA production in Alzheimer's disease. Mol Neurodegener 2025; 20:6. [PMID: 39815261 PMCID: PMC11734448 DOI: 10.1186/s13024-024-00788-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 12/05/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Alzheimer's Disease (AD) is a neurodegenerative disease with drastically altered astrocytic metabolism. Astrocytic GABA and H2O2 are associated with memory impairment in AD and synthesized through the Monoamine Oxidase B (MAOB)-mediated multi-step degradation of putrescine. However, the enzymes downstream to MAOB in this pathway remain unidentified. METHODS Using transcriptomics analysis, we identified two candidate enzymes, Aldehyde Dehydrogenase 1 family member A1 (ALDH1A1) and Sirtuin 2 (SIRT2) for the steps following MAOB in the astrocytic GABA production pathway. We used immunostaining, metabolite analysis and electrophysiology, both in vitro and in vivo, to confirm the participation of these enzymes in astrocytic GABA production. We checked for the presence of SIRT2 in human AD patients as well as the mouse model APP/PS1 and finally, we selectively ablated SIRT2 in the astrocytes of APP/PS1 mice to observe its effects on pathology. RESULTS Immunostaining, metabolite analysis, and electrophysiology recapitulated the participation of ALDH1A1 and SIRT2 in GABA production. Inhibition of SIRT2 reduced the production of astrocytic GABA but not H2O2, a key molecule in neurodegeneration. Elevated expression of these enzymes was found in hippocampal astrocytes of AD patients and APP/PS1 mice. Astrocyte-specific gene-silencing of SIRT2 in APP/PS1 mice restored GABA production and partially improved memory function. CONCLUSIONS Our study is the first to identify the specific role of SIRT2 in reactive astrogliosis and determine the specific pathway and metabolic step catalyzed by the enzyme. We determine the partial, yet significant role of ALDH1A1 in this process, thereby highlighting 2 new players the astrocytic GABA production pathway. Our findings therefore, offer SIRT2 as a new tool to segregate GABA from H2O2 production, aiding future research in neurodegenerative diseases.
Collapse
Affiliation(s)
- Mridula Bhalla
- Center for Cognition and Sociality, Life Science Institute (LSI), Institute for Basic Science (IBS), Daejeon, Republic of Korea
- IBS School, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Jinhyeong Joo
- Center for Cognition and Sociality, Life Science Institute (LSI), Institute for Basic Science (IBS), Daejeon, Republic of Korea
- IBS School, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Daeun Kim
- Center for Cognition and Sociality, Life Science Institute (LSI), Institute for Basic Science (IBS), Daejeon, Republic of Korea
- College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - Jeong Im Shin
- Center for Cognition and Sociality, Life Science Institute (LSI), Institute for Basic Science (IBS), Daejeon, Republic of Korea
- Division of Bio-Medical Science & Technology, Department of Neuroscience, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
| | - Yongmin Mason Park
- Center for Cognition and Sociality, Life Science Institute (LSI), Institute for Basic Science (IBS), Daejeon, Republic of Korea
- IBS School, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Yeon Ha Ju
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Uiyeol Park
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Seonguk Yoo
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Seung Jae Hyeon
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Hyunbeom Lee
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Junghee Lee
- Boston University Alzheimer's Disease Research Center and Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, USA.
- VA Boston Healthcare System, Boston, USA.
| | - Hoon Ryu
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - C Justin Lee
- Center for Cognition and Sociality, Life Science Institute (LSI), Institute for Basic Science (IBS), Daejeon, Republic of Korea.
- IBS School, University of Science and Technology (UST), Daejeon, Republic of Korea.
| |
Collapse
|
22
|
Rehman AS, Kumar P, Parvez S. Dopamine-D2-agonist targets mitochondrial dysfunction via diminishing Drp1 mediated fission and normalizing PGC1-α/SIRT3 pathways in a rodent model of Subarachnoid Haemorrhage. Neuroscience 2025; 564:60-78. [PMID: 39542343 DOI: 10.1016/j.neuroscience.2024.11.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/28/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
The adverse impact of disturbmitochondrialbiogenesis onearly brain injury (EBI) following subarachnoid haemorrhage (SAH) has been broadly recognized and is closely associated with oxidative stress and neuronal apoptosis. Previous studies have indicated the therapeutic potential of Ropinirole, a dopamine D2 agonist, in Ischemic Stroke. However, there is a lack of evidence regarding the ability of Ropinirole to enhance mitochondrial biogenesis and quality control after subarachnoid haemorrhage. The objective of this study is to investigate the effects of Ropinirole specific doses (10 & 20 mg/kg b. wt.) on mitochondria dysfunction in endovascular perforation SAH model in male Wistar rat. An endovascular perforation model was established using male Wistar rats that had sustained SAH injury. After the SAH injury, SAH grading on blood clot, Nissl staining, and neurobehavioral assessment were used to determine the severity. ROS and MMP, which are indicators of oxidative stress, were examined using flow cytometry. The findings demonstrated that the use of Ropinirole improved neurobehavioral outcomes, decreased brain edema, and reduced oxidative stress and mitochondrial based apoptosis. Further research showed that, Ropinirole therapy inhibit Drp1-mediated fission by accelerating the activity of fusion protein Mfn2/OPA1 along with regulating the translocation of PGC1-α and SIRT3 through restricting cytochrome C inside mitochondria to maintain mitochondrial metabolism. Ropinirole exerted neuroprotective effects by improving mitochondrial activity in a PGC1-α/SIRT3-dependent way via regulating Drp1 mediated fission. The effective treatment for SAH-induced EBI may involve increasing biogenesis and inhibiting excessive mitochondrial fission with Ropinirole.
Collapse
Affiliation(s)
- Ahmed Shaney Rehman
- Department of Medical Elementology & Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi 110042, India
| | - Suhel Parvez
- Department of Medical Elementology & Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
23
|
Mohan M, Mannan A, Nauriyal A, Singh TG. Emerging targets in amyotrophic lateral sclerosis (ALS): The promise of ATP-binding cassette (ABC) transporter modulation. Behav Brain Res 2025; 476:115242. [PMID: 39243983 DOI: 10.1016/j.bbr.2024.115242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative primarily affecting motor neurons, leading to disability and neuronal death, and ATP-Binding Cassette (ABC) transporter due to their role in drug efflux and modulation of various cellular pathways contributes to the pathogenesis of ALS. In this article, we extensively investigated various molecular and mechanistic pathways linking ALS transporter to the pathogenesis of ALS; this involves inflammatory pathways such as Mitogen-Activated Protein Kinase (MAPK), Phosphatidylinositol-3-Kinase/Protein Kinase B (PI3K/Akt), Toll-Like Receptor (TLR), Glycogen Synthase Kinase 3β (GSK-3β), Nuclear Factor Kappa-B (NF-κB), and Cyclooxygenase (COX). Oxidative pathways such as Astrocytes, Glutamate, Nuclear factor (erythroid-derived 2)-like 2 (Nrf2), Sirtuin 1 (SIRT-1), Forkhead box protein O (FOXO), Extracellular signal-regulated kinase (ERK). Additionally, we delve into the role of autophagic pathways like TAR DNA-binding protein 43 (TDP-43), AMP-activated protein kinase (AMPK), mammalian target of rapamycin (mTOR), and lastly, the apoptotic pathways. Furthermore, by understanding these intricate interactions, we aim to develop novel therapeutic strategies targeting ABC transporters, improving drug delivery, and ultimately offering a promising avenue for treating ALS.
Collapse
Affiliation(s)
- Maneesh Mohan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Aayush Nauriyal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
24
|
Zhou HL, Wang BB, Fan XL, Zhang XM, Song Y. Carvacrol acetate activated Nrf2 modulates mitophagy for the treatment of neurocyte oxidative stress induced by chlorpyrifos. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 289:117484. [PMID: 39644575 DOI: 10.1016/j.ecoenv.2024.117484] [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: 02/05/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
This study explored the protective effect and potential mechanism of carvacrol acetate (CAA) on the oxidation of chlorpyrifos (CPF). A model of oxidative stimulus damage was established in Sprague-Dawley rats by subcutaneous injection of the CPF poison. PC12 cells were used to construct an oxidative injury model using CPF, and the protective effects and mechanism of action of CAA against CPF-induced oxidative damage were explored in vitro. The key role of Nuclear factor erythroid-2-related factor 2 (Nrf2) in alleviating CPF-induced damage via CAA was further confirmed by administering Nrf2 inhibitors to PC12 cells. Administration of CAA significantly enhanced the locomotor ability of the rats, alleviated neuronal pathological alterations, and increased the number of Nissl bodies, while increasing autophagic bodies. In vitro, CAA promoted cell survival and augmented the mitochondrial membrane potential. It decreased both intra- and extracellular levels of reactive oxygen species (ROS), malondialdehyde (MDA), superoxide dismutase (SOD), while markedly elevating mitochondrial DNA (mtDNA) copy number. Moreover, PC12 cells treated with Nrf2 inhibitors failed to exhibit any improvement in survival rate when treated with CAA after a toxic insult. Furthermore, ROS and MDA levels were not significantly reduced, SOD enzyme activity did not increase, and mitochondrial membrane potential and mtDNA copy number did not improve. Western blot analysis showed that the expression of Tfam, Beclin1, and LC3II/LC3I proteins in the CAA group decreased significantly after Nrf2 inhibition. These findings suggest that CAA modulates mitochondrial function and autophagy by regulating the Nrf2 signalling pathway to mitigate the toxic damage. Finally, the effect of the autophagy inhibitor, 3-MA, on PC12 cells suggests that CAA promotes mitophagy by participating in the Nrf2 pathway, thereby preventing CPF-induced oxidative stress damage.
Collapse
Affiliation(s)
- Hong-Ling Zhou
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China.
| | - Bei-Bei Wang
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China.
| | - Xu-Li Fan
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China; Jinhua People's Hospital, Jinhua, Zhejiang 2321000, China.
| | - Xiao-Min Zhang
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China; First People's Hospital of Linping District, Hangzhou, Zhejiang 311103, China.
| | - Ying Song
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China; Hangzhou King's Bio-pharmaceutical Technology Co., Ltd., Hangzhou, Zhejiang 310007, China.
| |
Collapse
|
25
|
Tabassum S, Wu S, Lee CH, Yang BSK, Gusdon AM, Choi HA, Ren XS. Mitochondrial-targeted therapies in traumatic brain injury: From bench to bedside. Neurotherapeutics 2025; 22:e00515. [PMID: 39721917 PMCID: PMC11840356 DOI: 10.1016/j.neurot.2024.e00515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of morbidity and mortality worldwide, with limited effective therapeutic options currently available. Recent research has highlighted the pivotal role of mitochondrial dysfunction in the pathophysiology of TBI, making mitochondria an attractive target for therapeutic intervention. This review comprehensively examines advancements in mitochondrial-targeted therapies for TBI, bridging the gap from basic research to clinical applications. We discuss the underlying mechanisms of mitochondrial damage in TBI, including oxidative stress, impaired bioenergetics, mitochondrial dynamics, and apoptotic pathways. Furthermore, we highlight the complex interplay between mitochondrial dysfunction, inflammation, and blood-brain barrier (BBB) integrity, elucidating how these interactions exacerbate injury and impede recovery. We also evaluate various preclinical studies exploring pharmacological agents, gene therapy, and novel drug delivery systems designed to protect and restore mitochondrial function. Clinical trials and their outcomes are assessed to evaluate the translational potential of mitochondrial-targeted therapies in TBI. By integrating findings from bench to bedside, this review emphasizes promising therapeutic avenues and addresses remaining challenges. It also provides guidance for future research to pave the way for innovative treatments that improve patient outcomes in TBI.
Collapse
Affiliation(s)
- Sidra Tabassum
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Silin Wu
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Chang-Hun Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Bosco Seong Kyu Yang
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Aaron M Gusdon
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Huimahn A Choi
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Xuefang S Ren
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA.
| |
Collapse
|
26
|
Rawani NS, Chan AW, Todd KG, Baker GB, Dursun SM. The Role of Neuroglia in the Development and Progression of Schizophrenia. Biomolecules 2024; 15:10. [PMID: 39858403 PMCID: PMC11761573 DOI: 10.3390/biom15010010] [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: 09/25/2024] [Revised: 12/04/2024] [Accepted: 12/20/2024] [Indexed: 01/27/2025] Open
Abstract
Schizophrenia is a complex heterogenous disorder thought to be caused by interactions between genetic and environmental factors. The theories developed to explain the etiology of schizophrenia have focused largely on the dysfunction of neurotransmitters such as dopamine, serotonin and glutamate with their receptors, although research in the past several decades has indicated strongly that other factors are also involved and that the role of neuroglial cells in psychotic disorders including schizophrenia should be given more attention. Although glia were originally thought to be present in the brain only to support neurons in a physical, metabolic and nutritional capacity, it has become apparent that these cells have a variety of important physiological roles and that abnormalities in their function may make significant contributions to the symptoms of schizophrenia. In the present paper, we review the interactions of brain microglia, astrocytes and oligodendroglia with aspects such as transmitter dysregulation, neuro-inflammation, oxidative stress, synaptic function, the gut microbiome, myelination and the blood-brain barrier that appear to affect the cause, development and treatment of schizophrenia. We also review crosstalk between microglia, astrocytes and oligodendrocytes and the effects of antipsychotics on neuroglia. Problems associated with studies on specific biomarkers for glia in schizophrenia are discussed.
Collapse
Affiliation(s)
| | | | | | - Glen B. Baker
- Neurochemical Research Unit and Bebensee Schizophrenia Research Unit, Department of Psychiatry and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2G3, Canada; (N.S.R.); (A.W.C.); (K.G.T.); (S.M.D.)
| | | |
Collapse
|
27
|
Kaufmann WE, Luu S, Budimirovic DB. Drug Treatments for Neurodevelopmental Disorders: Targeting Signaling Pathways and Homeostasis. Curr Neurol Neurosci Rep 2024; 25:7. [PMID: 39641900 DOI: 10.1007/s11910-024-01394-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2024] [Indexed: 12/07/2024]
Abstract
PURPOSE OF THE REVIEW Preclinical and clinical evidence support the notion that neurodevelopmental disorders (NDDs) are synaptic disorders, characterized by excitatory-inhibitory imbalance. Despite this, NDD drug development programs targeting glutamate or gamma-aminobutyric acid (GABA) receptors have been largely unsuccessful. Nonetheless, recent drug trials in Rett syndrome (RTT), fragile X syndrome (FXS), and other NDDs targeting other mechanisms have met their endpoints. The purpose of this review is to identify the basis of these successful studies. RECENT FINDINGS Despite increasing evidence of disruption in synaptic homeostasis, most genetic variants associated with NDDs implicate proteins involved in cell regulation and not in neurotransmission. Metabolic processes, in particular mitochondrial function, appear to play a role in NDD pathophysiology. NDDs are also characterized by distinctive cell signaling abnormalities, which link cellular and synaptic homeostasis. Recent successful trials in NDDs, including those of trofinetide, the first drug specifically approved for one of these disorders (i.e., RTT), implicate the targeting of downstream processes (i.e., signaling pathways) rather than neurotransmitter receptors. Recent positive drug studies in NDDs and their underlying mechanisms, in conjunction with new knowledge on the pathophysiology of these disorders, support the concept that targeting signaling and cellular and synaptic homeostasis may be a preferred approach for ameliorating synaptic abnormalities in many NDDs.
Collapse
Affiliation(s)
- Walter E Kaufmann
- Boston Children's Hospital, Boston, MA, 02115, USA.
- Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Skylar Luu
- Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Dejan B Budimirovic
- Kennedy Krieger Institute and Department of Psychiatry & Behavioral Sciences-Child Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
28
|
Sámano C, Mazzone GL. The role of astrocytes response triggered by hyperglycaemia during spinal cord injury. Arch Physiol Biochem 2024; 130:724-741. [PMID: 37798949 DOI: 10.1080/13813455.2023.2264538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023]
Abstract
OBJECTIVE This manuscript aimed to provide a comprehensive overview of the physiological, molecular, and cellular mechanisms triggered by reactive astrocytes (RA) in the context of spinal cord injury (SCI), with a particular focus on cases involving hyperglycaemia. METHODS The compilation of articles related to astrocyte responses in neuropathological conditions, with a specific emphasis on those related to SCI and hyperglycaemia, was conducted by searching through databases including Science Direct, Web of Science, and PubMed. RESULTS AND CONCLUSIONS This article explores the dual role of astrocytes in both neurophysiological and neurodegenerative conditions within the central nervous system (CNS). In the aftermath of SCI and hyperglycaemia, astrocytes undergo a transformation into RA, adopting a distinct phenotype. While there are currently no approved therapies for SCI, various therapeutic strategies have been proposed to alleviate the detrimental effects of RAs following SCI and hyperglycemia. These strategies show promising potential in the treatment of SCI and its likely comorbidities.
Collapse
Affiliation(s)
- C Sámano
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana, Unidad Cuajimalpa (UAM-C), Ciudad de México, México
| | - G L Mazzone
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Pilar, Buenos Aires, Argentina
- Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Buenos Aires, Argentina
| |
Collapse
|
29
|
Khodir SA, Sweed EM, Faried MA, Abo Elkhair DM, Khalil MM, Afifi KH, El Agamy DF. Neuroprotective Effect of Maresin-1 in Rotenone-Induced Parkinson's Disease in Rats: The Putative Role of the JAK/STAT Pathway. Neurochem Res 2024; 50:30. [PMID: 39576344 PMCID: PMC11584474 DOI: 10.1007/s11064-024-04282-x] [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: 09/15/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/24/2024]
Abstract
Exposure to rotenone results in similar pathophysiological features as Parkinson's disease. Inflammation and oxidative stress are essential to PD pathogenesis. Maresin-1 has potent anti-inflammatory properties and promotes the regression of inflammation function. The current study aimed to evaluate the protective effects of Maresin-1 (MaR1) in rotenone (ROT)-induced PD and whether this protective role is associated with the initiation of the Janus kinase (JAK)-signal transducers and activator of transcription (STAT) signaling pathway. Thirty male Wister rats were classified into control, ROT-treated, and ROT + MaR1-treated groups. Rats underwent rotarod, open field, grip strength, and stepping tests as part of their motor behavioral evaluation. Serum glial cell-derived neurotrophic factor (GDNF) and striatal dopamine, acetylcholine, malondialdehyde (MDA), reduced glutathione (GSH), TNF-α, IL-6, and IL-1β were evaluated. Expression of JAK1 and STAT3 genes was assessed in striatum. Then, the tissue was subjected to histological and immunohistochemical evaluation for caspase-3, GFAP, and NF-kB. The administrated group with rotenone showed significant motor behavioral impairment. This was accompanied by reduced levels of GDNF and dopamine and increased levels of acetylcholine, as well as augmented oxidative stress and inflammatory biomarkers and reduced antioxidant activity. Inflammatory pathways (JAK1/STAT3, caspase-3, and NF-kB) were upregulated. Histopathological changes and upregulation in GFAP immunopositive reaction were observed. Remarkably, MaR1 treatment effectively alleviated behavior, histopathological changes, and biochemical alterations induced by ROT. MaR1 exerts protective effects against ROT-induced PD by its anti-inflammatory, antiapoptotic, and antioxidant properties. MaR1 mechanisms of action may involve modulation of pathways such as JAK/STAT.
Collapse
Affiliation(s)
- Suzan A Khodir
- Medical Physiology Department, Faculty of Medicine, Menoufia University, Menoufia, 32511, Egypt
- Medical Physiology Department, Menoufia National University, Menoufia, Egypt
| | - Eman M Sweed
- Clinical Pharmacology Department, Faculty of Medicine, Menoufia University, Menoufia, 32511, Egypt.
- Quality Assurance Center, Menoufia National University, Menoufia, Egypt.
| | - Manar A Faried
- Anatomy and Embryology Department, Faculty of Medicine, Menoufia University, Menoufia, 32511, Egypt
| | - Doaa M Abo Elkhair
- Anatomy and Embryology Department, Faculty of Medicine, Menoufia University, Menoufia, 32511, Egypt
| | - Marwa M Khalil
- Medical biochemistry and molecular biology Department, Faculty of Medicine, Menoufia University, Menoufia, 32511, Egypt
- Medical biochemistry and molecular biology Department, Menoufia National University, Menoufia, Egypt
| | - Khaled Hatem Afifi
- Neurology Department, Faculty of Medicine, Menoufia University, Menoufia, 32511, Egypt
| | - Dalia Fathy El Agamy
- Medical Physiology Department, Faculty of Medicine, Menoufia University, Menoufia, 32511, Egypt
- Medical Physiology Department, Menoufia National University, Menoufia, Egypt
| |
Collapse
|
30
|
Wong-Guerra M, Montano-Peguero Y, Ramírez-Sánchez J, Alfonso EG, Hernández-Enseñat D, Isaac YA, Padrón-Yaquis AS, da Rocha JBT, Fonseca-Fonseca LA, Núñez-Figueredo Y. Effect of JM-20 on Age-Related Cognitive Impairment in Mice. Neurochem Res 2024; 50:8. [PMID: 39546064 DOI: 10.1007/s11064-024-04254-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/05/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024]
Abstract
The decline in cognitive function associated with aging significantly impacts the well-being of elderly individuals and their families. This decline is a major recognized risk factor for neurodegenerative diseases, notably Alzheimer's disease. Animal models of aging provide a platform for evaluating drugs concerning aspects like memory and oxidative stress. JM-20 has demonstrated protective effects on short-term memory acquisition and consolidation, along with antioxidant properties and modulation of Acetylcholinesterase activity. This study assesses the potential protective JM-20 against cognitive decline and age-related memory loss. For the study, aged mice exhibiting aging-associated damage were initially selected. Experimental groups were then formed, and the effect of 8 mg/kg of JM-20 was evaluated for 40 days on aging-related behavior, such as spatial memory, novelty recognition memory, ambulatory activity, and anxiety. Subsequently, animals were sacrificed, and the hippocampal region was extracted for redox studies and to assess acetylcholinesterase activity. Results indicated that JM-20 at 8 mg/kg reversed damage to spatial working and reference memory, exhibiting performance comparable to untreated young adult animals. Furthermore, JM-20 preserved the enzymatic activity of superoxide dismutase, catalase, and total sulfhydryl levels in age-related cognitive impairment in mice, indicating a potent protective effect against oxidative events at the brain level. However, only young, healthy animals showed decreased acetylcholinesterase enzyme activity. These findings provide preclinical pharmacological evidence supporting the neuroprotective activity of JM-20, positioning it as a promising therapeutic candidate for treating memory disorders associated with aging.
Collapse
Affiliation(s)
- Maylin Wong-Guerra
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos (CIDEM), Ave 26 No. 1605 Boyeros y Puentes Grandes, La Habana, 10600, Cuba
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, USACH, Santiago, Alameda, 3363, Chile
| | - Yanay Montano-Peguero
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos (CIDEM), Ave 26 No. 1605 Boyeros y Puentes Grandes, La Habana, 10600, Cuba
- Facultad de Ciencias Químicas y Farmacéuticas, Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santos Dumont 964, Casilla 233, Santiago, Chile
| | - Jeney Ramírez-Sánchez
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos (CIDEM), Ave 26 No. 1605 Boyeros y Puentes Grandes, La Habana, 10600, Cuba
| | - Enrique García Alfonso
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos (CIDEM), Ave 26 No. 1605 Boyeros y Puentes Grandes, La Habana, 10600, Cuba
| | - Daniela Hernández-Enseñat
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos (CIDEM), Ave 26 No. 1605 Boyeros y Puentes Grandes, La Habana, 10600, Cuba
| | - Yeniceis Alcántara Isaac
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos (CIDEM), Ave 26 No. 1605 Boyeros y Puentes Grandes, La Habana, 10600, Cuba
| | - Alejandro Saúl Padrón-Yaquis
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos (CIDEM), Ave 26 No. 1605 Boyeros y Puentes Grandes, La Habana, 10600, Cuba
| | - João Batista Teixeira da Rocha
- Programa de Pós-graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria (UFSM), Avenida Roraima 1000, Santa Maria, RS, 97105-900, Brazil
| | - Luis Arturo Fonseca-Fonseca
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos (CIDEM), Ave 26 No. 1605 Boyeros y Puentes Grandes, La Habana, 10600, Cuba.
| | - Yanier Núñez-Figueredo
- Laboratorio de Neurofarmacología Experimental, Centro de Investigación y Desarrollo de Medicamentos (CIDEM), Ave 26 No. 1605 Boyeros y Puentes Grandes, La Habana, 10600, Cuba.
| |
Collapse
|
31
|
Patel MD, Lavekar SS, Jaisalmeria R, Oji S, Jayasi J, Cvetkovic C, Krencik R. Human Astrocytes Synchronize Neural Organoid Networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.17.618921. [PMID: 39464076 PMCID: PMC11507866 DOI: 10.1101/2024.10.17.618921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Biological neural networks exhibit synchronized activity within and across interconnected regions of the central nervous system. Understanding how these coordinated networks are established and maintained may reveal therapeutic targets for neurodegeneration and neuromodulation. Here, we tested the influence of astrocytes upon synchronous network activity using human pluripotent stem cell-derived bioengineered neural organoids. This study revealed that astrocytes significantly increase activity within individual organoids and across long distances among numerous rapidly merged organoids via influencing synapses and bioenergetics. Treatment of amyloid protein inhibited synchronous activity during neurodegeneration, yet this can be rescued by propagating activity from neighboring networks. Altogether, this study identifies critical contributions of human astrocytes to biological neural networks and delivers a rapid, reproducible, and scalable model to investigate long-range functional communication of the nervous system in healthy and disease states.
Collapse
|
32
|
Fanlo-Ucar H, Picón-Pagès P, Herrera-Fernández V, ILL-Raga G, Muñoz FJ. The Dual Role of Amyloid Beta-Peptide in Oxidative Stress and Inflammation: Unveiling Their Connections in Alzheimer's Disease Etiopathology. Antioxidants (Basel) 2024; 13:1208. [PMID: 39456461 PMCID: PMC11505517 DOI: 10.3390/antiox13101208] [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: 08/07/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/28/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease, and it is currently the seventh leading cause of death worldwide. It is characterized by the extracellular aggregation of the amyloid β-peptide (Aβ) into oligomers and fibrils that cause synaptotoxicity and neuronal death. Aβ exhibits a dual role in promoting oxidative stress and inflammation. This review aims to unravel the intricate connection between these processes and their contribution to AD progression. The review delves into oxidative stress in AD, focusing on the involvement of metals, mitochondrial dysfunction, and biomolecule oxidation. The distinct yet overlapping concept of nitro-oxidative stress is also discussed, detailing the roles of nitric oxide, mitochondrial perturbations, and their cumulative impact on Aβ production and neurotoxicity. Inflammation is examined through astroglia and microglia function, elucidating their response to Aβ and their contribution to oxidative stress within the AD brain. The blood-brain barrier and oligodendrocytes are also considered in the context of AD pathophysiology. We also review current diagnostic methodologies and emerging therapeutic strategies aimed at mitigating oxidative stress and inflammation, thereby offering potential treatments for halting or slowing AD progression. This comprehensive synthesis underscores the pivotal role of Aβ in bridging oxidative stress and inflammation, advancing our understanding of AD and informing future research and treatment paradigms.
Collapse
Affiliation(s)
- Hugo Fanlo-Ucar
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| | - Pol Picón-Pagès
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
- Laboratory of Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08028 Barcelona, Spain
| | - Víctor Herrera-Fernández
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| | - Gerard ILL-Raga
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| | - Francisco J. Muñoz
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| |
Collapse
|
33
|
Aliaghaei A, Meftahi GH. Silymarin ameliorates motor function and averts neuroinflammation-induced cell death in the rat model of Huntington's disease. Brain Res Bull 2024; 216:111039. [PMID: 39089590 DOI: 10.1016/j.brainresbull.2024.111039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/20/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
Huntington's disease (HD) is a scarce neurodegenerative disorder defined by chorea (unusual involuntary movements), behavioral presentations, psychiatric features, and cognitive deterioration. Although the precise pathogenic mechanism behind HD has not yet been identified, the most widely acknowledged pathways include excitotoxicity, mitochondrial malfunction, neuroinflammation, neurochemical imbalance, oxidative stress, and apoptosis HD has no efficient therapy. Current medications have drawbacks. Silymarin, a compound made up of standardized extracts obtained from the seeds of the Silybum marianum and polyphenolic flavonolignan, is utilized in therapeutic settings to treat a variety of experimental disorders in animals. Silymarin's key pharmacological activities include anti-cancer, hepatoprotection, antioxidant, cardioprotection, and anti-inflammatory. It also has no adverse side effects on people or animals. The current study aims to provide Silymarin's neuro-pharmacological activities or therapeutic qualities in HD. In this study, Thirty-six male Sprague-Dawley rats (200-220 g, 8 weeks) at the initial of the study were used. Silymarin solution (100 mg/Kg) was administered by oral gavage for 21 days to ameliorate neural damage in rats injected with 3-nitropropionicacid (3-NP) in a preliminary rat model of HD. The results showed that administration of silymarin to HD rats reduced gliosis, improved motor coordination and muscle activity, and increased striatal volume and the number of neurons and glial cells. Our results suggest that silymarin provides a protective environment for nerve cells and can have beneficial effects against the harmful effects of HD.
Collapse
Affiliation(s)
- Abbas Aliaghaei
- Hearing Disorders Research Center, Loghman‑Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gholam Hossein Meftahi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
34
|
Geng Y, Lou J, Wu J, Tao Z, Yang N, Kuang J, Wu Y, Zhang J, Xiang L, Shi J, Cai Y, Wang X, Chen J, Xiao J, Zhou K. NEMO-Binding Domain/IKKγ Inhibitory Peptide Alleviates Neuronal Pyroptosis in Spinal Cord Injury by Inhibiting ASMase-Induced Lysosome Membrane Permeabilization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405759. [PMID: 39225315 PMCID: PMC11516130 DOI: 10.1002/advs.202405759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 08/16/2024] [Indexed: 09/04/2024]
Abstract
A short peptide termed NEMO-binding domain (NBD) peptide has an inhibitory effect on nuclear factor kappa-B (NF-κB). Despite its efficacy in inhibiting inflammatory responses, the precise neuroprotective mechanisms of NBD peptide in spinal cord injury (SCI) remain unclear. This study aims to determine whether the pyroptosis-related aspects involved in the neuroprotective effects of NBD peptide post-SCI.Using RNA sequencing, the molecular mechanisms of NBD peptide in SCI are explored. The evaluation of functional recovery is performed using the Basso mouse scale, Nissl staining, footprint analysis, Masson's trichrome staining, and HE staining. Western blotting, enzyme-linked immunosorbent assays, and immunofluorescence assays are used to examine pyroptosis, autophagy, lysosomal membrane permeabilization (LMP), acid sphingomyelinase (ASMase), and the NF-κB/p38-MAPK related signaling pathway.NBD peptide mitigated glial scar formation, reduced motor neuron death, and enhanced functional recovery in SCI mice. Additionally, NBD peptide inhibits pyroptosis, ameliorate LMP-induced autophagy flux disorder in neuron post-SCI. Mechanistically, NBD peptide alleviates LMP and subsequently enhances autophagy by inhibiting ASMase through the NF-κB/p38-MAPK/Elk-1/Egr-1 signaling cascade, thereby mitigating neuronal death. NBD peptide contributes to functional restoration by suppressing ASMase-mediated LMP and autophagy depression, and inhibiting pyroptosis in neuron following SCI, which may have potential clinical application value.
Collapse
Affiliation(s)
- Yibo Geng
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Zhejiang Provincial Key Laboratory of OrthopaedicsWenzhou325027China
| | - Junsheng Lou
- Department of Orthopedic SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Junnan Wu
- Department of PharmacyThe Quzhou Affiliated Hospital of Wenzhou Medical UniversityQuzhou People's HospitalQuzhou324000China
| | - Zhichao Tao
- Renji College of Wenzhou Medical UniversityWenzhou325027China
| | - Ningning Yang
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Zhejiang Provincial Key Laboratory of OrthopaedicsWenzhou325027China
| | - Jiaxuan Kuang
- Cixi Biomedical Research InstituteWenzhou Medical UniversityNingbo315300China
| | - Yuzhe Wu
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Zhejiang Provincial Key Laboratory of OrthopaedicsWenzhou325027China
| | - Jiacheng Zhang
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Zhejiang Provincial Key Laboratory of OrthopaedicsWenzhou325027China
| | - Linyi Xiang
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Zhejiang Provincial Key Laboratory of OrthopaedicsWenzhou325027China
| | - Jingwei Shi
- Cixi Biomedical Research InstituteWenzhou Medical UniversityNingbo315300China
| | - Yuepiao Cai
- Cixi Biomedical Research InstituteWenzhou Medical UniversityNingbo315300China
- Molecular Pharmacology Research CenterSchool of Pharmaceutical ScienceWenzhou Medical UniversityWenzhou325027China
| | - Xiangyang Wang
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Zhejiang Provincial Key Laboratory of OrthopaedicsWenzhou325027China
| | - Jiaoxiang Chen
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Zhejiang Provincial Key Laboratory of OrthopaedicsWenzhou325027China
| | - Jian Xiao
- Cixi Biomedical Research InstituteWenzhou Medical UniversityNingbo315300China
- Molecular Pharmacology Research CenterSchool of Pharmaceutical ScienceWenzhou Medical UniversityWenzhou325027China
| | - Kailiang Zhou
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Zhejiang Provincial Key Laboratory of OrthopaedicsWenzhou325027China
- Cixi Biomedical Research InstituteWenzhou Medical UniversityNingbo315300China
| |
Collapse
|
35
|
Yassine M, Hassan SA, Yücel LA, Purath FFA, Korf HW, von Gall C, Ali AAH. Hepatocellular Carcinoma in Mice Affects Neuronal Activity and Glia Cells in the Suprachiasmatic Nucleus. Biomedicines 2024; 12:2202. [PMID: 39457515 PMCID: PMC11504045 DOI: 10.3390/biomedicines12102202] [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: 08/29/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Chronic liver diseases such as hepatic tumors can affect the brain through the liver-brain axis, leading to neurotransmitter dysregulation and behavioral changes. Cancer patients suffer from fatigue, which can be associated with sleep disturbances. Sleep is regulated via two interlocked mechanisms: homeostatic regulation and the circadian system. In mammals, the hypothalamic suprachiasmatic nucleus (SCN) is the key component of the circadian system. It generates circadian rhythms in physiology and behavior and controls their entrainment to the surrounding light/dark cycle. Neuron-glia interactions are crucial for the functional integrity of the SCN. Under pathological conditions, oxidative stress can compromise these interactions and thus circadian timekeeping and entrainment. To date, little is known about the impact of peripheral pathologies such as hepatocellular carcinoma (HCC) on SCN. Materials and Methods: In this study, HCC was induced in adult male mice. The key neuropeptides (vasoactive intestinal peptide: VIP, arginine vasopressin: AVP), an essential component of the molecular clockwork (Bmal1), markers for activity of neurons (c-Fos), astrocytes (GFAP), microglia (IBA1), as well as oxidative stress (8-OHdG) in the SCN were analyzed by immunohistochemistry at four different time points in HCC-bearing compared to control mice. Results: The immunoreactions for VIP, Bmal1, GFAP, IBA1, and 8-OHdG were increased in HCC mice compared to control mice, especially during the activity phase. In contrast, c-Fos was decreased in HCC mice, especially during the late inactive phase. Conclusions: Our data suggest that HCC affects the circadian system at the level of SCN. This involves an alteration of neuropeptides, neuronal activity, Bmal1, activation of glia cells, and oxidative stress in the SCN.
Collapse
Affiliation(s)
- Mona Yassine
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany; (M.Y.); (S.A.H.); (L.A.Y.); (F.F.A.P.); (A.A.H.A.)
| | - Soha A. Hassan
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany; (M.Y.); (S.A.H.); (L.A.Y.); (F.F.A.P.); (A.A.H.A.)
- Department of Zoology, Faculty of Science, Suez University, P.O. Box 43221, Suez 43533, Egypt
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Lea Aylin Yücel
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany; (M.Y.); (S.A.H.); (L.A.Y.); (F.F.A.P.); (A.A.H.A.)
| | - Fathima Faiba A. Purath
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany; (M.Y.); (S.A.H.); (L.A.Y.); (F.F.A.P.); (A.A.H.A.)
| | - Horst-Werner Korf
- Institute of Anatomy I, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany;
| | - Charlotte von Gall
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany; (M.Y.); (S.A.H.); (L.A.Y.); (F.F.A.P.); (A.A.H.A.)
| | - Amira A. H. Ali
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany; (M.Y.); (S.A.H.); (L.A.Y.); (F.F.A.P.); (A.A.H.A.)
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
36
|
Boonpraman N, Yi SS. NADPH oxidase 4 (NOX4) as a biomarker and therapeutic target in neurodegenerative diseases. Neural Regen Res 2024; 19:1961-1966. [PMID: 38227522 DOI: 10.4103/1673-5374.390973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/25/2023] [Indexed: 01/17/2024] Open
Abstract
Diseases like Alzheimer's and Parkinson's diseases are defined by inflammation and the damage neurons undergo due to oxidative stress. A primary reactive oxygen species contributor in the central nervous system, NADPH oxidase 4, is viewed as a potential therapeutic touchstone and indicative marker for these ailments. This in-depth review brings to light distinct features of NADPH oxidase 4, responsible for generating superoxide and hydrogen peroxide, emphasizing its pivotal role in activating glial cells, inciting inflammation, and disturbing neuronal functions. Significantly, malfunctioning astrocytes, forming the majority in the central nervous system, play a part in advancing neurodegenerative diseases, due to their reactive oxygen species and inflammatory factor secretion. Our study reveals that aiming at NADPH oxidase 4 within astrocytes could be a viable treatment pathway to reduce oxidative damage and halt neurodegenerative processes. Adjusting NADPH oxidase 4 activity might influence the neuroinflammatory cytokine levels, including myeloperoxidase and osteopontin, offering better prospects for conditions like Alzheimer's disease and Parkinson's disease. This review sheds light on the role of NADPH oxidase 4 in neural degeneration, emphasizing its drug target potential, and paving the path for novel treatment approaches to combat these severe conditions.
Collapse
Affiliation(s)
- Napissara Boonpraman
- BK21 four Program, Department of Medical Sciences, Soonchunhyang University, Asan, South Korea
| | - Sun Shin Yi
- BK21 four Program, Department of Medical Sciences, Soonchunhyang University, Asan, South Korea
- Department of Biomedical Laboratory Science, Soonchunhyang University, Asan, South Korea
- iConnectome, Co., Ltd., Cheonan, South Korea
| |
Collapse
|
37
|
Wang Z, Zhang X, Zhang G, Zheng YJ, Zhao A, Jiang X, Gan J. Astrocyte modulation in cerebral ischemia-reperfusion injury: A promising therapeutic strategy. Exp Neurol 2024; 378:114814. [PMID: 38762094 DOI: 10.1016/j.expneurol.2024.114814] [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: 01/25/2024] [Revised: 04/03/2024] [Accepted: 05/12/2024] [Indexed: 05/20/2024]
Abstract
Cerebral ischemia-reperfusion injury (CIRI) poses significant challenges for drug development due to its complex pathogenesis. Astrocyte involvement in CIRI pathogenesis has led to the development of novel astrocyte-targeting drug strategies. To comprehensively review the current literature, we conducted a thorough analysis from January 2012 to December 2023, identifying 82 drugs aimed at preventing and treating CIRI. These drugs target astrocytes to exert potential benefits in CIRI, and their primary actions include modulation of relevant signaling pathways to inhibit neuroinflammation and oxidative stress, reduce cerebral edema, restore blood-brain barrier integrity, suppress excitotoxicity, and regulate autophagy. Notably, active components from traditional Chinese medicines (TCM) such as Salvia miltiorrhiza, Ginkgo, and Ginseng exhibit these important pharmacological properties and show promise in the treatment of CIRI. This review highlights the potential of astrocyte-targeted drugs to ameliorate CIRI and categorizes them based on their mechanisms of action, underscoring their therapeutic potential in targeting astrocytes.
Collapse
Affiliation(s)
- Ziyu Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaolu Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guangming Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yu Jia Zheng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Anliu Zhao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Jiali Gan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
38
|
Ogunsuyi OB, Ogunruku OO, Umar HI, Oboh G. Effect of curcumin-donepezil combination on spatial memory, astrocyte activation, and cholinesterase expressions in brain of scopolamine-treated rats. Mol Biol Rep 2024; 51:864. [PMID: 39073463 DOI: 10.1007/s11033-024-09712-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 06/06/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND The study investigated the effect of co-administration of curcumin and donepezil on several markers of cognitive function (such as spatial memory, astrocyte activation, cholinesterase expressions) in the brain cortex and hippocampus of scopolamine-treated rats. METHOD AND RESULTS For seven consecutive days, a pre-treatment of curcumin (50 mg/kg) and/or donepezil (2.5 mg/kg) was administered. On the seventh day, scopolamine (1 mg/kg) was administered to elicit cognitive impairment, 30 min before memory test was conducted. This was followed by evaluating changes in spatial memory, cholinesterase, and adenosine deaminase (ADA) activities, as well as nitric oxide (NO) level were determined. Additionally, RT-qPCR for glial fibrillary acidic protein (GFAP) and cholinesterase gene expressions was performed in the brain cortex and hippocampus. Also, GFAP immunohistochemistry of the brain tissues for neuronal injury were performed in the brain cortex and hippocampus. In comparison to the control group, rats given scopolamine had impaired memory, higher levels of acetylcholinesterase (AChE), butyrylcholinesterase (BChE), and ADA activities, as well as elevated markers of oxidative stress. In addition to enhanced GFAP immunoreactivity, there was also overexpression of the GFAP and BChE genes in the brain tissues. The combination of curcumin and donepezil was, however, observed to better ameliorate these impairments in comparison to the donepezil-administered rat group. CONCLUSION Hence, this evidence provides more mechanisms to support the hypothesis that the concurrent administration of curcumin and donepezil mitigates markers of cognitive dysfunction in scopolamine-treated rat model.
Collapse
Affiliation(s)
- Opeyemi Babatunde Ogunsuyi
- Department of Biomedical Technology, School of Basic Medical Sciences, The Federal University of Technology, Akure, Nigeria.
- Drosophila Research Lab, Functional Foods and Nutraceuticals Unit, The Federal University of Technology, Akure, Nigeria.
| | | | - Haruna Isiyaku Umar
- Department of Biochemistry, The Federal University of Technology, Akure, Nigeria
- Molecular Biology and Bioinformatics Lab, Department of Biochemistry, The Federal University of Technology, Akure, Nigeria
| | - Ganiyu Oboh
- Drosophila Research Lab, Functional Foods and Nutraceuticals Unit, The Federal University of Technology, Akure, Nigeria
- Department of Biochemistry, The Federal University of Technology, Akure, Nigeria
| |
Collapse
|
39
|
Oginga FO, Mpofana T. Understanding the role of early life stress and schizophrenia on anxiety and depressive like outcomes: An experimental study. Behav Brain Res 2024; 470:115053. [PMID: 38768688 DOI: 10.1016/j.bbr.2024.115053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND Adverse experiences due to early life stress (ELS) or parental psychopathology such as schizophrenia (SZ) have a significant implication on individual susceptibility to psychiatric disorders in the future. However, it is not fully understood how ELS affects social-associated behaviors as well as the developing prefrontal cortex (PFC). OBJECTIVE The aim of this study was to investigate the impact of ELS and ketamine induced schizophrenia like symptoms (KSZ) on anhedonia, social behavior and anxiety-like behavior. METHODS Male and female Sprague-Dawley rat pups were allocated randomly into eight experimental groups, namely control, gestational stress (GS), GS+KSZ, maternal separation (MS), MS+KSZ pups, KSZ parents, KSZ parents and Pups and KSZ pups only. ELS was induced by subjecting the pups to GS and MS, while schizophrenia like symptoms was induced through subcutaneous administration of ketamine. Behavioral assessment included sucrose preference test (SPT) and elevated plus maze (EPM), followed by dopamine testing and analysis of astrocyte density. Statistical analysis involved ANOVA and post hoc Tukey tests, revealing significant group differences and yielding insights into behavioral and neurodevelopmental impacts. RESULTS GS, MS, and KSZ (dams) significantly reduced hedonic response and increased anxiety-like responses (p < 0.05). Notably, the presence of normal parental mental health demonstrated a reversal of the observed decline in Glial Fibrillary Acidic Protein-positive astrocytes (GFAP+ astrocytes) (p < 0.05) and a reduction in anxiety levels, implying its potential protective influence on depressive-like symptoms and PFC astrocyte functionality. CONCLUSION The present study provides empirical evidence supporting the hypothesis that exposure to ELS and KSZ on dams have a significant impact on the on development of anxiety and depressive like symptoms in Sprague Dawley rats, while positive parenting has a reversal effect.
Collapse
Affiliation(s)
- Fredrick Otieno Oginga
- Department of Physiology, School of Laboratory Medicine and Medical Sciences, University of Kwa-Zulu Natal, Durban 4001, South Africa; Department of Clinical Medicine, School of Medicine and Health Science, Kabarak University, Nakuru 20157, Kenya.
| | - Thabisile Mpofana
- Department of Human Physiology, Faculty of Health Sciences North West University, Potchefstroom campus, 11 Hoffman St., Potchefstroom 2531, South Africa
| |
Collapse
|
40
|
Sirajo MU, Maigari YK, Sunusi A, Jibril AN, Lawal IU, Ibrahim BM. Synergistic action of vitamin D3 and A on motor activity regulation in mice model of extrapyramidal syndrome: Correlational insights into astrocyte regulation, cytokine modulation, and dopaminergic activity. J Chem Neuroanat 2024; 138:102421. [PMID: 38649035 DOI: 10.1016/j.jchemneu.2024.102421] [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: 01/10/2024] [Revised: 04/05/2024] [Accepted: 04/13/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Extrapyramidal syndromes (EPS) represent neurological side effects of antipsychotic medications, characterized by motor disturbances. While previous studies have indicated the neuroprotective effects of vitamin D and A against EPS, the underlying mechanisms of this protection remain unclear. METHODS Twenty-four adult mice were categorized into four groups: positive and negative control groups, one receiving a dopamine antagonist, and the other receiving both a dopamine antagonist and vitamins D and A. Sections of the corticobasal loop, specifically the motor cortex (M1) and basal nuclei (CPu), were prepared for Immunohistochemistry (IHC) and stained with Glial Fibrillary Acidic Protein (GFAP) to visualize reactive astrocytes. ELISA assays for TNF-α, IL-6, IL-4, IL-13, and dopamine levels were performed on homogenized brain sections. RESULTS The EPS group exhibited a significant increase in TNF-α and IL-6 levels in M1 and CPu. Treatment with dopamine agonists and vitamin D&A resulted in significant reductions in IL-6 levels. Only the Vitamin D&A group showed a significant decline in TNF-α. The EPS group recorded significant decreases in IL-4 and IL-13, with IL-13 significantly elevated in the dopamine agonist and Vitamin D&A groups. IL-4 was notably increased in the Vitamin D&A groups. Dopamine concentration significantly declined in the EPS group, with improvements observed in the groups treated with dopamine agonists, and vitamin D&A. Reactive astrocytes were significantly expressed in the M1 and CPu of the EPS group but poorly expressed in other groups. CONCLUSIONS EPS is linked to astrocyte activation, an upsurge in pro-inflammatory cytokines, a decline in anti-inflammatory cytokines, and dopamine in the corticobasal loop. Administration of vitamin D3 and A was found to suppres pro-inflammatory cytokines and repress anti-inflammatory cytokines associated with astrocyte activation.
Collapse
Affiliation(s)
- Mujittapha U Sirajo
- Anatomy Unit, Department of Physiotherapy, School of Basic Medical Sciences, Skyline University Nigeria, Kano, Nigeria; Department of Anatomy, College of Health Sciences, Bayero University Kano, Nigeria
| | - Yahya K Maigari
- Department of Anatomy, College of Health Sciences, Bayero University Kano, Nigeria
| | - Abdulrashid Sunusi
- Department of Anatomy, College of Health Sciences, Bayero University Kano, Nigeria
| | - Adam N Jibril
- Department of Anatomy, College of Health Sciences, Bayero University Kano, Nigeria
| | - Isa Usman Lawal
- Anatomy Unit, Department of Physiotherapy, School of Basic Medical Sciences, Skyline University Nigeria, Kano, Nigeria; Department of Physiotherapy, Faculty of Allied Health Sciences, College of Health Sciences, Bayero University Kano, Nigeria
| | - Badamasi M Ibrahim
- Anatomy Unit, Department of Physiotherapy, School of Basic Medical Sciences, Skyline University Nigeria, Kano, Nigeria; Department of Anatomy, College of Health Sciences, Bayero University Kano, Nigeria.
| |
Collapse
|
41
|
Alhadidi QM, Bahader GA, Arvola O, Kitchen P, Shah ZA, Salman MM. Astrocytes in functional recovery following central nervous system injuries. J Physiol 2024; 602:3069-3096. [PMID: 37702572 PMCID: PMC11421637 DOI: 10.1113/jp284197] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/07/2023] [Indexed: 09/14/2023] Open
Abstract
Astrocytes are increasingly recognised as partaking in complex homeostatic mechanisms critical for regulating neuronal plasticity following central nervous system (CNS) insults. Ischaemic stroke and traumatic brain injury are associated with high rates of disability and mortality. Depending on the context and type of injury, reactive astrocytes respond with diverse morphological, proliferative and functional changes collectively known as astrogliosis, which results in both pathogenic and protective effects. There is a large body of research on the negative consequences of astrogliosis following brain injuries. There is also growing interest in how astrogliosis might in some contexts be protective and help to limit the spread of the injury. However, little is known about how astrocytes contribute to the chronic functional recovery phase following traumatic and ischaemic brain insults. In this review, we explore the protective functions of astrocytes in various aspects of secondary brain injury such as oedema, inflammation and blood-brain barrier dysfunction. We also discuss the current knowledge on astrocyte contribution to tissue regeneration, including angiogenesis, neurogenesis, synaptogenesis, dendrogenesis and axogenesis. Finally, we discuss diverse astrocyte-related factors that, if selectively targeted, could form the basis of astrocyte-targeted therapeutic strategies to better address currently untreatable CNS disorders.
Collapse
Affiliation(s)
- Qasim M Alhadidi
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Pharmacy, Al-Yarmok University College, Diyala, Iraq
| | - Ghaith A Bahader
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Oiva Arvola
- Division of Anaesthesiology, Jorvi Hospital, Department of Anaesthesiology, Intensive Care and Pain Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Philip Kitchen
- College of Health and Life Sciences, Aston University, Birmingham, UK
| | - Zahoor A Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Mootaz M Salman
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Kavli Institute for NanoScience Discovery, University of Oxford, Oxford, UK
| |
Collapse
|
42
|
Pietrucha A, Serdar M, Bendix I, Endesfelder S, Brinke EAD, Urkola A, Bührer C, Schmitz T, Scheuer T. Oxygen and HIF1α-dependent SDF1 expression in primary astrocytes. Dev Neurobiol 2024; 84:113-127. [PMID: 38544386 DOI: 10.1002/dneu.22938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 07/17/2024]
Abstract
In the naturally hypoxic in utero fetal environment of preterm infants, oxygen and oxygen-sensitive signaling pathways play an important role in brain development, with hypoxia-inducible factor-1α (HIF1α) being an important regulator. Early exposure to nonphysiological high oxygen concentrations by birth in room can induce HIF1α degradation and may affect neuronal and glial development. This involves the dysregulation of astroglial maturation and function, which in turn might contribute to oxygen-induced brain injury. In this study, we investigated the effects of early high oxygen exposure on astroglial maturation and, specifically, on astroglial stromal cell-derived factor 1 (SDF1) expression in vivo and in vitro. In our neonatal mouse model of hyperoxia preterm birth brain injury in vivo, high oxygen exposure affected astroglial development and cortical SDF1 expression. These results were further supported by reduced Sdf1 expression, impaired proliferation, decreased total cell number, and altered expression of astroglial markers in astrocytes in primary cultures grown under high oxygen conditions. Moreover, to mimic the naturally hypoxic in utero fetal environment, astroglial Sdf1 expression was increased after low oxygen exposure in vitro, which appears to be regulated by HIF1α activity. Additionally, the knockdown of Hif1α revealed HIF1α-dependent Sdf1 expression in vitro. Our results indicate HIF1α and oxygen-dependent chemokine expression in primary astrocytes and highlight the importance of oxygen conditions for brain development.
Collapse
Affiliation(s)
- Andreas Pietrucha
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Meray Serdar
- Department of Pediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational and Behavioral Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ivo Bendix
- Department of Pediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational and Behavioral Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Stefanie Endesfelder
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Elena Auf dem Brinke
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Ane Urkola
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Christoph Bührer
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Thomas Schmitz
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Till Scheuer
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| |
Collapse
|
43
|
Sun M, Chen Z. Unveiling the Complex Role of Exosomes in Alzheimer's Disease. J Inflamm Res 2024; 17:3921-3948. [PMID: 38911990 PMCID: PMC11193473 DOI: 10.2147/jir.s466821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/11/2024] [Indexed: 06/25/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative illness, characterized by memory loss and cognitive decline, accounting for 60-80% of dementia cases. AD is characterized by senile plaques made up of amyloid β (Aβ) protein, intracellular neurofibrillary tangles caused by hyperphosphorylation of tau protein linked with microtubules, and neuronal loss. Currently, therapeutic treatments and nanotechnological developments are effective in treating the symptoms of AD, but a cure for the illness has not yet been found. Recently, the increased study of extracellular vesicles (EVs) has led to a growing awareness of their significant involvement in neurodegenerative disorders, including AD. Exosomes are small extracellular vesicles that transport various components including messenger RNAs, non-coding RNAs, proteins, lipids, DNA, and other bioactive compounds from one cell to another, facilitating information transmission and material movement. There is growing evidence indicating that exosomes have complex functions in AD. Exosomes may have a dual role in Alzheimer's disease by contributing to neuronal death and also helping to alleviate the pathological progression of the disease. Therefore, the primary aim of this review is to outline the updated understandings on exosomes biogenesis and many functions of exosomes in the generation, conveyance, distribution, and elimination of hazardous proteins related to Alzheimer's disease. This review is intended to provide novel insights for understanding the development, specific treatment, and early detection of Alzheimer's disease.
Collapse
Affiliation(s)
- Mingyue Sun
- Department of Neurology, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Zhuoyou Chen
- Department of Neurology, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| |
Collapse
|
44
|
Rawani NS, Chan AW, Dursun SM, Baker GB. The Underlying Neurobiological Mechanisms of Psychosis: Focus on Neurotransmission Dysregulation, Neuroinflammation, Oxidative Stress, and Mitochondrial Dysfunction. Antioxidants (Basel) 2024; 13:709. [PMID: 38929148 PMCID: PMC11200831 DOI: 10.3390/antiox13060709] [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: 03/25/2024] [Revised: 05/16/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Psychosis, defined as a set of symptoms that results in a distorted sense of reality, is observed in several psychiatric disorders in addition to schizophrenia. This paper reviews the literature relevant to the underlying neurobiology of psychosis. The dopamine hypothesis has been a major influence in the study of the neurochemistry of psychosis and in development of antipsychotic drugs. However, it became clear early on that other factors must be involved in the dysfunction involved in psychosis. In the current review, it is reported how several of these factors, namely dysregulation of neurotransmitters [dopamine, serotonin, glutamate, and γ-aminobutyric acid (GABA)], neuroinflammation, glia (microglia, astrocytes, and oligodendrocytes), the hypothalamic-pituitary-adrenal axis, the gut microbiome, oxidative stress, and mitochondrial dysfunction contribute to psychosis and interact with one another. Research on psychosis has increased knowledge of the complexity of psychotic disorders. Potential new pharmacotherapies, including combinations of drugs (with pre- and probiotics in some cases) affecting several of the factors mentioned above, have been suggested. Similarly, several putative biomarkers, particularly those related to the immune system, have been proposed. Future research on both pharmacotherapy and biomarkers will require better-designed studies conducted on an all stages of psychotic disorders and must consider confounders such as sex differences and comorbidity.
Collapse
Affiliation(s)
| | | | | | - Glen B. Baker
- Neurochemical Research Unit and Bebensee Schizophrenia Research Unit, Department of Psychiatry and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2G3, Canada; (N.S.R.); (A.W.C.); (S.M.D.)
| |
Collapse
|
45
|
Stanton AE, Bubnys A, Agbas E, James B, Park DS, Jiang A, Pinals RL, Liu L, Truong N, Loon A, Staab C, Cerit O, Wen HL, Kellis M, Blanchard JW, Langer R, Tsai LH. Engineered 3D Immuno-Glial-Neurovascular Human miBrain Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.15.553453. [PMID: 37645757 PMCID: PMC10461996 DOI: 10.1101/2023.08.15.553453] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Patient-specific, human-based cellular models integrating a biomimetic blood-brain barrier (BBB), immune, and myelinated neuron components are critically needed to enable accelerated, translationally relevant discovery of neurological disease mechanisms and interventions. By engineering a novel brain-mimicking 3D hydrogel and co-culturing all six major brain cell types derived from patient iPSCs, we have constructed, characterized, and utilized a multicellular integrated brain (miBrain) immuno-glial-neurovascular model with in vivo- like hallmarks inclusive of neuronal activity, functional connectivity, barrier function, myelin-producing oligodendrocyte engagement with neurons, multicellular interactions, and transcriptomic profiles. We implemented the model to study Alzheimer's Disease pathologies associated with APOE4 genetic risk. APOE4 miBrains differentially exhibit amyloid aggregation, tau phosphorylation, and astrocytic GFAP. Unlike the co-emergent fate specification of glia and neurons in organoids, miBrains integrate independently differentiated cell types, a feature we harnessed to identify that APOE4 in astrocytes promotes neuronal tau pathogenesis and dysregulation through crosstalk with microglia.
Collapse
|
46
|
Hidalgo-Lanussa O, González Santos J, Barreto GE. Sex-specific vulnerabilities in human astrocytes underpin the differential impact of palmitic acid. Neurobiol Dis 2024; 195:106489. [PMID: 38552721 DOI: 10.1016/j.nbd.2024.106489] [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: 02/21/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/05/2024] Open
Abstract
Obesity and neurometabolic diseases have been linked to neurodegenerative diseases. Our hypothesis is that the endogenous estrogenic component of human astrocytes plays a critical role in cell response during lipotoxic damage, given that obesity can disrupt hormonal homeostasis and cause brain inflammation. Our findings showed that high concentrations of palmitic acid (PA) significantly reduced cell viability more in male astrocytes, indicating sex-specific vulnerabilities. PA induced a greater increase in cytosolic reactive oxygen species (ROS) production in males, while female astrocytes exhibited higher superoxide ion levels in mitochondria. In addition, female astrocytes treated with PA showed increased expression of antioxidant proteins, including catalase, Gpx-1 and Nrf2 suggesting a stronger cellular defence mechanism. Interestingly, there was a difference in the expression of estrogenic components, such as estrogen, androgens, and progesterone receptors, as well as aromatase and 5α-reductase enzymes, between males and females. PA induced their expression mainly in females, indicating a potential protective mechanism mediated by endogenous hormones. In summary, our findings highlight the impact of sex on the response of human astrocytes to lipotoxicity. Male astrocytes appear to be more susceptible to cellular damage when exposed to high concentrations of fatty acids.
Collapse
Affiliation(s)
- Oscar Hidalgo-Lanussa
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia; Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - Janneth González Santos
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.
| |
Collapse
|
47
|
Shuvalova M, Dmitrieva A, Belousov V, Nosov G. The role of reactive oxygen species in the regulation of the blood-brain barrier. Tissue Barriers 2024:2361202. [PMID: 38808582 DOI: 10.1080/21688370.2024.2361202] [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] [Accepted: 05/23/2024] [Indexed: 05/30/2024] Open
Abstract
The blood-brain barrier (BBB) regulates the exchange of metabolites and cells between the blood and brain, and maintains central nervous system homeostasis. Various factors affect BBB barrier functions, including reactive oxygen species (ROS). ROS can act as stressors, damaging biological molecules, but they also serve as secondary messengers in intracellular signaling cascades during redox signaling. The impact of ROS on the BBB has been observed in multiple sclerosis, stroke, trauma, and other neurological disorders, making blocking ROS generation a promising therapeutic strategy for BBB dysfunction. However, it is important to consider ROS generation during normal BBB functioning for signaling purposes. This review summarizes data on proteins expressed by BBB cells that can be targets of redox signaling or oxidative stress. It also provides examples of signaling molecules whose impact may cause ROS generation in the BBB, as well as discusses the most common diseases associated with BBB dysfunction and excessive ROS generation, open questions that arise in the study of this problem, and possible ways to overcome them.
Collapse
Affiliation(s)
- Margarita Shuvalova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of metabolism and redox biology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Anastasiia Dmitrieva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Vsevolod Belousov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of metabolism and redox biology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Skolkovo, Moscow, Russia
| | - Georgii Nosov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Skolkovo, Moscow, Russia
| |
Collapse
|
48
|
Pandya VA, Patani R. The role of glial cells in amyotrophic lateral sclerosis. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 176:381-450. [PMID: 38802179 DOI: 10.1016/bs.irn.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) has traditionally been considered a neuron-centric disease. This view is now outdated, with increasing recognition of cell autonomous and non-cell autonomous contributions of central and peripheral nervous system glia to ALS pathomechanisms. With glial research rapidly accelerating, we comprehensively interrogate the roles of astrocytes, microglia, oligodendrocytes, ependymal cells, Schwann cells and satellite glia in nervous system physiology and ALS-associated pathology. Moreover, we highlight the inter-glial, glial-neuronal and inter-system polylogue which constitutes the healthy nervous system and destabilises in disease. We also propose classification based on function for complex glial reactive phenotypes and discuss the pre-requisite for integrative modelling to advance translation. Given the paucity of life-enhancing therapies currently available for ALS patients, we discuss the promising potential of harnessing glia in driving ALS therapeutic discovery.
Collapse
Affiliation(s)
- Virenkumar A Pandya
- University College London Medical School, London, United Kingdom; The Francis Crick Institute, London, United Kingdom.
| | - Rickie Patani
- The Francis Crick Institute, London, United Kingdom; Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, Queen Square, London, United Kingdom.
| |
Collapse
|
49
|
Liang Y, Shi Y, Guo R, Xu C, Fu M, Shen J, Gao X, Li W, Qin K. Wine- and stir-frying processing of Cuscutae Semen enhance its ability to alleviate oxidative stress and apoptosis via the Keap 1-Nrf2/HO-1 and PI3K/AKT pathways in H 2O 2-challenged KGN human granulosa cell line. BMC Complement Med Ther 2024; 24:189. [PMID: 38750475 PMCID: PMC11094956 DOI: 10.1186/s12906-024-04491-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/07/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Cuscutae Semen (CS) has been prescribed in traditional Chinese medicine (TCM) for millennia as an aging inhibitor, an anti-inflammatory agent, a pain reliever, and an aphrodisiac. Its three main forms include crude Cuscutae Semen (CCS), wine-processed CS (WCS), and stir-frying-processed CS (SFCS). Premature ovarian insufficiency (POI) is a globally occurring medical condition. The present work sought a highly efficacious multi-target therapeutic approach against POI with minimal side effects. Finally, it analyzed the relative differences among CCS, WCS and SFCS in terms of their therapeutic efficacy and modes of action against H2O2-challenged KGN human granulosa cell line. METHODS In this study, ultrahigh-performance liquid chromatography (UPLC)-Q-ExactiveTM Orbitrap-mass spectrometry (MS), oxidative stress indices, reactive oxygen species (ROS), Mitochondrial membrane potential (MMP), real-time PCR, Western blotting, and molecular docking were used to investigate the protective effect of CCS, WCS and SFCS on KGN cells oxidative stress and apoptosis mechanisms. RESULTS The results confirmed that pretreatment with CCS, WCS and SFCS reduced H2O2-induced oxidative damage, accompanied by declining ROS levels and malondialdehyde (MDA) accumulation in the KGN cells. CCS, WCS and SFCS upregulated the expression of antioxidative levels (GSH, GSH/GSSG ratio, SOD, T-AOC),mitochondrial membrane potential (MMP) and the relative mRNA(Nrf2, Keap1, NQO-1, HO-1, SOD-1, CAT). They inhibited apoptosis by upregulating Bcl-2, downregulating Bax, cleaved caspase-9, and cleaved caspase-3, and lowering the Bax/Bcl-2 ratio. They also exerted antioxidant efficacy by partially activating the PI3K/Akt and Keap1-Nrf2/HO-1 signaling pathways. CONCLUSIONS The results of the present work demonstrated the inhibitory efficacy of CCS, WCS and SFCS against H2O2-induced oxidative stress and apoptosis in KGN cells and showed that the associated mechanisms included Keap1-Nrf2/HO-1 activation, P-PI3K upregulation, and P-Akt-mediated PI3K-Akt pathway induction.
Collapse
Affiliation(s)
- Yusha Liang
- School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yun Shi
- School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Rong Guo
- School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Changli Xu
- Department of Pharmacy, Xinyi People's Hospital, Xinyi Jiangsu, 221400, China
| | - Mian Fu
- School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jinyang Shen
- School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Xun Gao
- School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| | - Weidong Li
- Engineering Research Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Kunming Qin
- School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| |
Collapse
|
50
|
Lysikova T, Tomascova A, Kovalska M, Lehotsky J, Leskova Majdova K, Kaplan P, Tatarkova Z. Dynamics in Redox-Active Molecules Following Ischemic Preconditioning in the Brain. Neurol Int 2024; 16:533-550. [PMID: 38804479 PMCID: PMC11130914 DOI: 10.3390/neurolint16030040] [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: 03/25/2024] [Revised: 04/28/2024] [Accepted: 05/06/2024] [Indexed: 05/29/2024] Open
Abstract
It is well known that the brain is quite vulnerable to oxidative stress, initiating neuronal loss after ischemia-reperfusion (IR) injury. A potent protective mechanism is ischemic preconditioning (IPC), where proteins are among the primary targets. This study explores redox-active proteins' role in preserving energy supply. Adult rats were divided into the control, IR, and IPC groups. Protein profiling was conducted to identify modified proteins and then verified through activity assays, immunoblot, and immunohistochemical analyses. IPC protected cortex mitochondria, as evidenced by a 2.26-fold increase in superoxide dismutase (SOD) activity. Additionally, stable core subunits of respiratory chain complexes ensured sufficient energy production, supported by a 16.6% increase in ATP synthase activity. In hippocampal cells, IPC led to the downregulation of energy-related dehydrogenases, while a significantly higher level of peroxiredoxin 6 (PRX6) was observed. Notably, IPC significantly enhanced glutathione reductase activity to provide sufficient glutathione to maintain PRX6 function. Astrocytes may mobilize PRX6 to protect neurons during initial ischemic events, by decreased PRX6 positivity in astrocytes, accompanied by an increase in neurons following both IR injury and IPC. Maintained redox signaling via astrocyte-neuron communication triggers IPC's protective state. The partnership among PRX6, SOD, and glutathione reductase appears essential in safeguarding and stabilizing the hippocampus.
Collapse
Affiliation(s)
- Terezia Lysikova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (T.L.); (A.T.); (J.L.); (K.L.M.); (P.K.)
| | - Anna Tomascova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (T.L.); (A.T.); (J.L.); (K.L.M.); (P.K.)
| | - Maria Kovalska
- Department of Histology and Embryology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Jan Lehotsky
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (T.L.); (A.T.); (J.L.); (K.L.M.); (P.K.)
| | - Katarina Leskova Majdova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (T.L.); (A.T.); (J.L.); (K.L.M.); (P.K.)
| | - Peter Kaplan
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (T.L.); (A.T.); (J.L.); (K.L.M.); (P.K.)
| | - Zuzana Tatarkova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (T.L.); (A.T.); (J.L.); (K.L.M.); (P.K.)
| |
Collapse
|