51
|
Xu J, Yan Z, Bang S, Velmeshev D, Ji RR. GPR37L1 identifies spinal cord astrocytes and protects neuropathic pain after nerve injury. Neuron 2025; 113:1206-1222.e6. [PMID: 39952243 PMCID: PMC12005970 DOI: 10.1016/j.neuron.2025.01.012] [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: 03/23/2024] [Revised: 09/10/2024] [Accepted: 01/16/2025] [Indexed: 02/17/2025]
Abstract
Astrocytes in the spinal cord dorsal horn (SDH) play a pivotal role in synaptic transmission and neuropathic pain. However, the precise classification of SDH astrocytes in health and disease remains elusive. Here, we reveal Gpr37l1 as a marker and functional regulator of spinal astrocytes. Through single-nucleus RNA sequencing, we identified Gpr37l1 as a selective G-protein-coupled receptor (GPCR) marker for spinal cord astrocytes. Notably, SDH displayed reactive astrocyte phenotypes and exacerbated neuropathic pain following nerve injury combined with Gpr37l1 deficiency. In naive animals, Gpr37l1 knockdown in SDH astrocytes induces astrogliosis and pain hypersensitivity, while Gpr37l1-/- mice fail to recover from neuropathic pain. GPR37L1 activation by maresin 1 increased astrocyte glutamate transporter 1 (GLT-1) activity and reduced spinal EPSCs and neuropathic pain. Selective overexpression of Gpr37l1 in SDH astrocytes reversed neuropathic pain and astrogliosis after nerve injury. Our findings illuminate astrocyte GPR37l1 as an essential negative regulator of pain, which protects against neuropathic pain through astrocyte signaling in SDH.
Collapse
Affiliation(s)
- Jing Xu
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Zihan Yan
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Sangsu Bang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Dmitry Velmeshev
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
52
|
Wu SR, Nowakowski TJ. Exploring human brain development and disease using assembloids. Neuron 2025; 113:1133-1150. [PMID: 40107269 PMCID: PMC12022838 DOI: 10.1016/j.neuron.2025.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 01/10/2025] [Accepted: 02/12/2025] [Indexed: 03/22/2025]
Abstract
How the human brain develops and what goes awry in neurological disorders represent two long-lasting questions in neuroscience. Owing to the limited access to primary human brain tissue, insights into these questions have been largely gained through animal models. However, there are fundamental differences between developing mouse and human brain, and neural organoids derived from human pluripotent stem cells (hPSCs) have recently emerged as a robust experimental system that mimics self-organizing and multicellular features of early human brain development. Controlled integration of multiple organoids into assembloids has begun to unravel principles of cell-cell interactions. Moreover, patient-derived or genetically engineered hPSCs provide opportunities to investigate phenotypic correlates of neurodevelopmental disorders and to develop therapeutic hypotheses. Here, we outline the advances in technologies that facilitate studies by using assembloids and summarize their applications in brain development and disease modeling. Lastly, we discuss the major roadblocks of the current system and potential solutions.
Collapse
Affiliation(s)
- Sih-Rong Wu
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Tomasz J Nowakowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
53
|
Said N, Venketaraman V. Neuroinflammation, Blood-Brain Barrier, and HIV Reservoirs in the CNS: An In-Depth Exploration of Latency Mechanisms and Emerging Therapeutic Strategies. Viruses 2025; 17:572. [PMID: 40285014 PMCID: PMC12030944 DOI: 10.3390/v17040572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/12/2025] [Accepted: 04/12/2025] [Indexed: 04/29/2025] Open
Abstract
Despite the success of antiretroviral therapy (ART) in suppressing viral replication in the blood, HIV persists in the central nervous system (CNS) and causes chronic neurocognitive impairment, a hallmark of HIV-associated neurocognitive disorders (HAND). This review looks at the complex interactions among HIV, the blood-brain barrier (BBB), neuroinflammation, and the roles of viral proteins, immune cell trafficking, and pro-inflammatory mediators in establishing and maintaining latent viral reservoirs in the CNS, particularly microglia and astrocytes. Key findings show disruption of the BBB, monocyte infiltration, and activation of CNS-resident cells by HIV proteins like Tat and gp120, contributing to the neuroinflammatory environment and neuronal damage. Advances in epigenetic regulation of latency have identified targets like histone modifications and DNA methylation, and new therapeutic strategies like latency-reversing agents (LRAs), gene editing (CRISPR/Cas9), and nanoparticle-based drug delivery also offer hope. While we have made significant progress in understanding the molecular basis of HIV persistence in the CNS, overcoming the challenges of BBB penetration and neuroinflammation is key to developing effective therapies. Further research into combination therapies and novel drug delivery systems will help improve outcomes for HAND patients and bring us closer to a functional cure for HIV.
Collapse
Affiliation(s)
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766-1854, USA;
| |
Collapse
|
54
|
Gaweda-Walerych K, Aragona V, Lodato S, Sitek EJ, Narożańska E, Buratti E. Progranulin deficiency in the brain: the interplay between neuronal and non-neuronal cells. Transl Neurodegener 2025; 14:18. [PMID: 40234992 PMCID: PMC12001433 DOI: 10.1186/s40035-025-00475-8] [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: 09/20/2024] [Accepted: 02/21/2025] [Indexed: 04/17/2025] Open
Abstract
Heterozygous mutations in GRN gene lead to insufficient levels of the progranulin (PGRN) protein, resulting in frontotemporal dementia (FTD) with TAR DNA-binding protein 43 (TDP-43) inclusions, classified pathologically as frontotemporal lobar degeneration (FTLD-TDP). Homozygous GRN mutations are exceedingly rare and cause neuronal ceroid lipofuscinosis 11, a lysosomal storage disease with onset in young adulthood, or an FTD syndrome with late-onset manifestations. In this review, we highlight the broad spectrum of clinical phenotypes associated with PGRN deficiency, including primary progressive aphasia and behavioral variant of frontotemporal dementia. We explore these phenotypes alongside relevant rodent and in vitro human models, ranging from the induced pluripotent stem cell-derived neural progenitors, neurons, microglia, and astrocytes to genetically engineered heterotypic organoids containing both neurons and astrocytes. We summarize advantages and limitations of these models in recapitulating the main FTLD-GRN hallmarks, highlighting the role of non-cell-autonomous mechanisms in the formation of TDP-43 pathology, neuroinflammation, and neurodegeneration. Data obtained from patients' brain tissues and biofluids, in parallel with single-cell transcriptomics, demonstrate the complexity of interactions among the highly heterogeneous cellular clusters present in the brain, including neurons, astrocytes, microglia, oligodendroglia, endothelial cells, and pericytes. Emerging evidence has revealed that PGRN deficiency is associated with cell cluster-specific, often conserved, genetic and molecular phenotypes in the central nervous system. In this review, we focus on how these distinct cellular populations and their dysfunctional crosstalk contribute to neurodegeneration and neuroinflammation in FTD-GRN. Specifically, we characterize the phenotypes of lipid droplet-accumulating microglia and alterations of myelin lipid content resulting from lysosomal dysfunction caused by PGRN deficiency. Additionally, we consider how the deregulation of glia-neuron communication affects the exchange of organelles such as mitochondria, and the removal of excess toxic products such as protein aggregates, in PGRN-related neurodegeneration.
Collapse
Affiliation(s)
- Katarzyna Gaweda-Walerych
- Department of Neurogenetics and Functional Genomics, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland.
| | - Vanessa Aragona
- Department of Biomedical Sciences, Humanitas University, Via Levi Montalicini 4, Pieve Emanuele, 20072, Milan, Italy
- Neurodevelopment Biology Lab, IRCCS Humanitas Research Hospital, via Manzoni, 56, Rozzano, 20089, Milan, Italy
| | - Simona Lodato
- Department of Biomedical Sciences, Humanitas University, Via Levi Montalicini 4, Pieve Emanuele, 20072, Milan, Italy
- Neurodevelopment Biology Lab, IRCCS Humanitas Research Hospital, via Manzoni, 56, Rozzano, 20089, Milan, Italy
| | - Emilia J Sitek
- Division of Neurological and Psychiatric Nursing, Laboratory of Clinical Neuropsychology, Neurolinguistics, and Neuropsychotherapy, Faculty of Health Sciences, Medical University of Gdansk, 80-210, Gdansk, Poland.
- Neurology Department, St. Adalbert Hospital, Copernicus PL, 80-462, Gdansk, Poland.
| | - Ewa Narożańska
- Neurology Department, St. Adalbert Hospital, Copernicus PL, 80-462, Gdansk, Poland
| | - Emanuele Buratti
- Molecular Pathology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), AREA Science Park, 34149, Trieste, Italy
| |
Collapse
|
55
|
Kunze LH, Palumbo G, Gnörich J, Wind-Mark K, Schaefer R, Lindner S, Gildehaus FJ, Ziegler S, Brendel M. Fibrillar amyloidosis and synaptic vesicle protein expression progress jointly in the cortex of a mouse model with β-amyloid pathology. Neuroimage 2025; 310:121165. [PMID: 40120783 DOI: 10.1016/j.neuroimage.2025.121165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/13/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025] Open
Abstract
Neurodegeneration, accumulation of β-amyloid (Aβ) plaques, and neuroinflammation are the major hallmarks of Alzheimer's disease. Here, we aimed to investigate the temporal and spatial association between synaptic activity, Aβ plaque load, and neuroinflammation in an Aβ mouse model with limited neurodegeneration. 26 APPSL70 and 15 C57Bl/6 mice underwent longitudinal PET-scans with [18F]UCB-H from plaque onset to levels of strong plaque load (5.3 - 11.0 months of age) to assess the synaptic vesicle protein 2A (SV2A) expression, [18F]FBB to determine the fibrillar Aβ plaque load, and [18F]GE-180 and [18F]F-DED to assess microglial and astroglial (re)activity. Statistical parametric mapping was performed to uncover similarities between the binding patterns of all four tracers. We found a continuous increase in Aβ-PET in APPSL70 mice from 5.3 to 11.0 months of age, resulting in a significantly higher [18F]FBB PET signal in the cortex, hippocampus, and thalamus of APPSL70 mice compared to C57Bl/6 mice at 11.0 months of age. Parallel increases in SV2A-PET signals were observed in the cortex and thalamus of APPSL70 mice compared to C57Bl/6 mice. Statistical parametric mapping revealed a similar pattern of Aβ- and SV2A-PET differences (dice coefficient 53 %). Patterns of microglia activation showed stronger congruency with SV2A expression (dice coefficient 58 %) than patterns of reactive astrogliosis (dice coefficient 26 %). APPSL70 mice with limited neurodegeneration comprise a close temporal and spatial association between SV2A expression, Aβ plaque load, and microglial activation. SV2A PET imaging in APPSL70 mice may facilitate longitudinal monitoring of increased synaptic activity in the earliest phase of AD.
Collapse
Affiliation(s)
- L H Kunze
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - G Palumbo
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - J Gnörich
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - K Wind-Mark
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - R Schaefer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - S Lindner
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - F-J Gildehaus
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - S Ziegler
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany; Department of Nuclear Medicine, Hannover Medical School (MHH), Hannover, Germany
| | - M Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Ludwig Maximilian University of Munich, 81377 Munich, Germany.
| |
Collapse
|
56
|
Wang X, Zhou Z, Zhang Y, Liu J, Qin T, Zhou W, Li Q, Wu X, Xue K, Cao H, Su Y, Zhao S, Lu C, Jiang T, Yin G, Chen J. Exosome-shuttled miR-5121 from A2 astrocytes promotes BSCB repair after traumatic SCI by activating autophagy in vascular endothelial cells. J Nanobiotechnology 2025; 23:291. [PMID: 40229869 PMCID: PMC11998472 DOI: 10.1186/s12951-025-03365-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/01/2025] [Indexed: 04/16/2025] Open
Abstract
Spinal cord injury (SCI) is a severe neurological disorder that significantly impacts patients' quality of life. Following SCI, the blood-spinal cord barrier (BSCB) is destroyed, leading to ischemia and hypoxia, which further exacerbates the imbalance in the spinal cord microenvironment. A2-type astrocytes, which arise under ischemic and hypoxic conditions, have been reported to promote SCI repair. However, the roles of exosomes derived from A2 astrocytes (A2-Exos) in SCI have not been explored. This study aims to investigate the role of A2-Exos in SCI repair, particularly in BSCB restoration, and to elucidate its potential mechanisms. GEO database analysis, western blotting, and immunofluorescence were used to detect A2 astrocyte polarization after SCI in mice. In vitro, A2 astrocytes were obtained through hypoxia induction, and A2-Exos were extracted via ultracentrifugation. An in vivo SCI model and a series of in vitro experiments demonstrated the reparative effects of A2-Exos on BSCB following SCI. Furthermore, miRNA sequencing analysis and rescue experiments confirmed the role of miRNAs in A2-Exos-mediated BSCB repair. Finally, luciferase assays and western blotting were performed to investigate the underlying mechanisms. The results showed that A2-Exos promote motor function recovery and BSCB repair in mice following SCI. In vitro, A2-Exos facilitated BSCB reconstruction and endothelial cell autophagy. miRNA sequencing identified miR-5121 as the most significantly enriched miRNA in A2-Exos, suggesting its involvement in BSCB repair and autophagy regulation. AKT2 was identified as a potential downstream target of miR-5121. Functional gain- and loss-of-function experiments further validated the miR-5121/AKT2 axis. Finally, we demonstrated that the AKT2/mTOR/p70S6K pathway may mediate the effects of miR-5121 in A2-Exos on BSCB repair.
Collapse
Affiliation(s)
- Xiaowei Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
- Department of Orthopedics, Maanshan People's Hospital, Maanshan, Anhui, 243000, China
| | - Zihan Zhou
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Yu Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Jiayun Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Tao Qin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Wei Zhou
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Qingqing Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Xincan Wu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Kaixiao Xue
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Heng Cao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Yunxin Su
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Shujie Zhao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China
| | - Chun Lu
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, China.
| | - Tao Jiang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China.
| | - Guoyong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China.
| | - Jian Chen
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Jiangsu Provincial Clinical Research Institute, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|
57
|
Bettcher BM, de Oliveira FF, Willette AA, Michalowska MM, Machado LS, Rajbanshi B, Borelli WV, Tansey MG, Rocha A, Suryadevara V, Hu WT. Analysis and interpretation of inflammatory fluid markers in Alzheimer's disease: a roadmap for standardization. J Neuroinflammation 2025; 22:105. [PMID: 40234920 PMCID: PMC11998147 DOI: 10.1186/s12974-025-03432-4] [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: 01/17/2025] [Accepted: 03/31/2025] [Indexed: 04/17/2025] Open
Abstract
Growing interest in the role of the immune response in Alzheimer's Disease and related dementias (ADRD) has led to widespread use of fluid inflammatory markers in research studies. To standardize the use and interpretation of inflammatory markers in AD research, we build upon prior guidelines to develop consensus statements and recommendations to advance application and interpretation of these markers. In this roadmap paper, we propose a glossary of terms related to the immune response in the context of biomarker discovery/validation, discuss current conceptualizations of inflammatory markers in research, and recommend best practices to address key knowledge gaps. We also provide consensus principles to summarize primary conceptual, methodological, and interpretative issues facing the field: (1) a single inflammatory marker is likely insufficient to describe an entire biological cascade, and multiple markers with similar or distinct functions should be simultaneously measured in a panel; (2) association studies in humans are insufficient to infer causal relationships or mechanisms; (3) neuroinflammation displays time-dependent and disease context-dependent patterns; (4) neuroinflammatory mechanisms should not be inferred based solely on blood inflammatory marker changes; and (5) standardized reporting of CSF inflammatory marker assay validation and performance will improve incorporation of inflammatory markers into the biological AD criteria.
Collapse
Affiliation(s)
- Brianne M Bettcher
- Department of Neurology, University of Colorado Anschutz Medical Campus, 12469 East 17th Place, Room 217- Campus Box F429, Aurora, CO, 80045, USA.
| | | | - Auriel A Willette
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School and Center for Healthy Aging Research, Rutgers Institute for Health, Health Care Policy, and Aging Research, Rutgers Health, New Brunswick, USA
| | - Malgorzata M Michalowska
- Department of Clinical Neuroscience, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Luiza Santos Machado
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Binita Rajbanshi
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California - San Francisco, San Francisco, USA
| | - Wyllians V Borelli
- Department of Morphological Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Malú Gámez Tansey
- Department of Neurology, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, USA
| | - Andréia Rocha
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, USA
| | | | - William T Hu
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School and Center for Healthy Aging Research, Rutgers Institute for Health, Health Care Policy, and Aging Research, Rutgers Health, New Brunswick, USA
| |
Collapse
|
58
|
Ribeiro AR, Pereira R, Barros C, Barateiro A, Alberro A, Basto AP, Graça L, Pinto MV, Santos FMF, Gois PMP, Howlett SE, Fernandes A. Experimental autoimmune encephalomyelitis pathogenesis alters along animal age: impact of S100B expression. J Neuroimmune Pharmacol 2025; 20:37. [PMID: 40227512 PMCID: PMC11997003 DOI: 10.1007/s11481-025-10195-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/20/2025] [Indexed: 04/15/2025]
Abstract
Multiple Sclerosis (MS) is the leading inflammatory and non-traumatic cause of disability in young adults, with late-onset MS emerging in middle-aged patients often resulting in poorer treatment responses and worse prognoses. The calcium-binding protein S100B is elevated in MS patients, and its targeting has shown promise in reducing disease severity in experimental autoimmune encephalomyelitis (EAE) models. However, most studies on MS pathology have focused on young animal models, leaving a gap in understanding the effects of age and S100B ablation on disease progression throughout the lifespan. This study aimed to characterize EAE in mice of different ages, examining demyelination, inflammation, and immune responses to determine whether S100B ablation could mitigate MS pathogenesis across the lifespan. EAE was induced in six cohorts of C57BL/6 mice: young adults (3 months), older adults (6 months), and middle-aged (12 months), including corresponding S100B knockout (KO) groups, followed for 23 days. Upon sacrifice, spinal cords were assessed via immunohistochemistry and Real-Time qPCR, while splenocytes were analyzed for immune cell characterization. Results indicated a more severe disease course in 12-month-old mice, marked by increased gliosis, inflammation, and impaired microglial phagocytic activity. Notably, S100B absence reduced gliosis and inflammatory markers across all ages, with 12-month-old S100B KO mice showing increased regulatory T cells. These findings highlight the exacerbating role of age and elevated S100B in MS progression, underscoring the importance of identifying age-specific MS markers and therapeutic targets.
Collapse
MESH Headings
- Animals
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- S100 Calcium Binding Protein beta Subunit/genetics
- S100 Calcium Binding Protein beta Subunit/biosynthesis
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Female
- Aging/metabolism
- Aging/pathology
- Age Factors
- Spinal Cord/pathology
- Spinal Cord/metabolism
- Male
Collapse
Affiliation(s)
- Ana Rita Ribeiro
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Raquel Pereira
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Catarina Barros
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Andreia Barateiro
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
- Departamento de Ciências Farmacêuticas E Do Medicamento, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Ainhoa Alberro
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
- IIS Biogipuzkoa Health Research Institute, San Sebastian, Spain
| | - Afonso P Basto
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisbon, Portugal
- Laboratório Associado Para a Ciência Animal E Veterinária (AL4AnimalS), Lisbon, Portugal
| | - Luís Graça
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
- Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Vaz Pinto
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Fábio M F Santos
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Pedro M P Gois
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
- Departamento de Ciências Farmacêuticas E Do Medicamento, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Susan E Howlett
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
- Department of Medicine (Geriatric Medicine), Dalhousie University, Halifax, NS, Canada
| | - Adelaide Fernandes
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal.
- Departamento de Ciências Farmacêuticas E Do Medicamento, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
59
|
Wu M, Chen YF, Yao W, Zhou S, Xie Z, Tao Y, Zhong Y, Ma W. The anti-inflammatory drug Montelukast ameliorates cognitive deficits by rescuing the inflammatory levels in young AD animal models. Sci Rep 2025; 15:12720. [PMID: 40222965 PMCID: PMC11994820 DOI: 10.1038/s41598-025-91785-4] [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: 05/14/2024] [Accepted: 02/24/2025] [Indexed: 04/15/2025] Open
Abstract
Neuroinflammation precedes the clinical symptoms onset of Alzheimer's disease (AD) by decades. However, the anti-inflammatory drugs were not always effective at all stages of the disease. Here, using the fly and mouse AD models, we evaluated the effects of anti-inflammatory drugs on inflammatory-related factors and the proinflammatory cytokines at different ages of AD animals. We also performed behavioral tests to evaluate the cognitive aspects of AD. Combined with the bioinformatics analysis, we would like to exhibit a better understanding of AD. Based on the previous studies and reanalysis of published database, we found aged AD animals might better represent the inflammatory status of symptomatic AD. Our results showed that mRNA levels of antimicrobial peptides (AMPs) were highly expressed in 10-day-old AD flies, while no significant difference was observed in 40-day-old AD. In aged APP/PS1 mice (22.5 months), inflammatory-related factors NF-κB, IBA1, and the mRNA levels of proinflammatory cytokines Il-1β and Il-6 were not differentially expressed. In contrast, a significant increase was observed in 7.5-month-old APP/PS1 mice. Moreover, the anti-inflammatory drug Montelukast (MON) did not ameliorate the inflammatory and cognitive defects in 22.5-month-old aged mice but showed a rescue effect in 7.5-month-old young APP/PS1 mice. Altogether, our study demonstrates the different inflammatory status might lead to variations of anti-inflammatory drug efficacy, which helps to clarify the importance of considering the pathological stage of the disease when administering treatment.
Collapse
Affiliation(s)
- Mengnan Wu
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yan-Fen Chen
- Beijing Joekai Biotechnology LLC, Beijing, 100094, China
| | - Wei Yao
- Beijing Joekai Biotechnology LLC, Beijing, 100094, China
| | - Siyan Zhou
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Zuolei Xie
- Beijing Joekai Biotechnology LLC, Beijing, 100094, China
| | - Ye Tao
- Beijing Joekai Biotechnology LLC, Beijing, 100094, China
| | - Yi Zhong
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| | - Weiwei Ma
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Beijing Joekai Biotechnology LLC, Beijing, 100094, China.
| |
Collapse
|
60
|
Wen X, Cao W, Ding H, Chen A, Sun Z, Wang Y, Xi Y, Wu S. Extracellular Vesicles Derived from FGF2-Primed Astrocytes Against Mitochondrial and Synaptic Toxicities in Parkinson's Disease. Int J Nanomedicine 2025; 20:4627-4644. [PMID: 40248029 PMCID: PMC12005207 DOI: 10.2147/ijn.s511474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 04/07/2025] [Indexed: 04/19/2025] Open
Abstract
Purpose Mitochondrial dysfunction associated with neuronal degeneration and subsequent synaptic disconnection are essential for the development of Parkinson's disease (PD). Considering that astrocytes play key roles in synaptogenesis during development, we hypothesized that fibroblast growth factor - 2 (FGF2), a key factor for astrocyte development, could reverse the toxic phenotype of reactive astrocytes, and the extracellular vesicles (EVs) derived from FGF2-primed astrocytes would enhance synaptogenesis in PD model. The present study was to test this hypothesis. Methods EVs isolated from FGF2-primed astrocytes (FGF2-EVs) were characterized by transmission electron microscopy and nanoparticle tracking analysis. FGF2-EVs were applied to both in vitro and in vivo models of PD. EVs derived from naïve astrocytes (CON-EV) were used as control. Mitochondrial alterations, neuronal survival, synaptogenesis, and mice behavior were subsequently evaluated by quantitative real-time polymerase chain reaction, Western-blotting, immunohistochemistry, and CatWalk gait analysis. To dissect the underlying mechanisms, proteomic analysis and small interfering RNA (siRNA) mediated gene silencing were adopted. Results FGF2 treatment restored the expression of neural progenitor markers and suppressed the levels of A1 astrocytic markers in MPP+ pretreated astrocytes. FGF2-EVs, in comparison with that of CON-EVs, effectively protected neurons from mitochondrial fragmentation and stimulated synaptogenesis, as evidenced by expression of Mitofusin 2 (Mfn2), postsynaptic density protein 95 (PSD-95) and synaptophysin (SYP). Proteomic analysis revealed high enrichment of neural cell adhesion molecule 1 (NCAM1) in FGF2-EVs. Knocking down NCAM1 severely influenced the expression of mitochondrial and synaptic proteins. Furthermore, delivery of FGF2-EVs significantly enhanced the survival of TH+ neurons, the levels of NCAM1 and synaptogenesis in the substantia nigra of PD mice, as well as the locomotion of PD mice. Conclusion EVs from FGF2-primed astrocytes are superior in protecting PD mice against mitochondrial and synaptic toxicities, possibly through NCAM1, which could be used as a therapeutic strategy for PD.
Collapse
Affiliation(s)
- Xiaomin Wen
- College of Life Sciences, Northwest University, Xi’an, Shaanxi, 710069, People’s Republic of China
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Wanjun Cao
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Hui Ding
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Andi Chen
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Zhichuan Sun
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Yazhou Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Ye Xi
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Shengxi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| |
Collapse
|
61
|
Garton T, Smith MD, Kesharwani A, Gharagozloo M, Oh S, Na CH, Absinta M, Reich DS, Zack DJ, Calabresi PA. Myeloid lineage C3 induces reactive gliosis and neuronal stress during CNS inflammation. Nat Commun 2025; 16:3481. [PMID: 40216817 PMCID: PMC11992029 DOI: 10.1038/s41467-025-58708-3] [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: 08/04/2024] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Complement component C3 mediates pathology in CNS neurodegenerative diseases. Here we use scRNAseq of sorted C3-reporter positive cells from mouse brain and optic nerve to characterize C3 producing glia in experimental autoimmune encephalomyelitis (EAE), a model in which peripheral immune cells infiltrate the CNS, causing reactive gliosis and neuro-axonal pathology. We find that C3 expression in the early inflammatory stage of EAE defines disease-associated glial subtypes characterized by increased expression of genes associated with mTOR activation and cell metabolism. This pro-inflammatory subtype is abrogated with genetic C3 depletion, a finding confirmed with proteomic analyses. In addition, early optic nerve axonal injury and retinal ganglion cell oxidative stress, but not loss of post-synaptic density protein 95, are ameliorated by selective deletion of C3 in myeloid cells. These data suggest that in addition to C3b opsonization of post synaptic proteins leading to neuronal demise, C3 activation is a contributor to reactive glia in the optic nerve.
Collapse
Affiliation(s)
- Thomas Garton
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Matthew D Smith
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Ajay Kesharwani
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Marjan Gharagozloo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Sungtaek Oh
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Chan-Hyun Na
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Martina Absinta
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- IRCCS Humanitas Reserach Hospital, Milan, Italy
| | - Daniel S Reich
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Donald J Zack
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
62
|
D'Sa K, Choi ML, Wagen AZ, Setó-Salvia N, Kopach O, Evans JR, Rodrigues M, Lopez-Garcia P, Lachica J, Clarke BE, Singh J, Ghareeb A, Bayne J, Grant-Peters M, Garcia-Ruiz S, Chen Z, Rodriques S, Athauda D, Gustavsson EK, Gagliano Taliun SA, Toomey C, Reynolds RH, Young G, Strohbuecker S, Warner T, Rusakov DA, Patani R, Bryant C, Klenerman DA, Gandhi S, Ryten M. Astrocytic RNA editing regulates the host immune response to alpha-synuclein. SCIENCE ADVANCES 2025; 11:eadp8504. [PMID: 40215316 PMCID: PMC11988446 DOI: 10.1126/sciadv.adp8504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 03/07/2025] [Indexed: 04/14/2025]
Abstract
RNA editing is a posttranscriptional mechanism that targets changes in RNA transcripts to modulate innate immune responses. We report the role of astrocyte-specific, ADAR1-mediated RNA editing in neuroinflammation in Parkinson's disease (PD). We generated human induced pluripotent stem cell-derived astrocytes, neurons and cocultures and exposed them to small soluble alpha-synuclein aggregates. Oligomeric alpha-synuclein triggered an inflammatory glial state associated with Toll-like receptor activation, viral responses, and cytokine secretion. This reactive state resulted in loss of neurosupportive functions and the induction of neuronal toxicity. Notably, interferon response pathways were activated leading to up-regulation and isoform switching of the RNA deaminase enzyme, ADAR1. ADAR1 mediates A-to-I RNA editing, and increases in RNA editing were observed in inflammatory pathways in cells, as well as in postmortem human PD brain. Aberrant, or dysregulated, ADAR1 responses and RNA editing may lead to sustained inflammatory reactive states in astrocytes triggered by alpha-synuclein aggregation, and this may drive the neuroinflammatory cascade in Parkinson's.
Collapse
Affiliation(s)
- Karishma D'Sa
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Minee L. Choi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Brain & Cognitive Sciences, KAIST, 921 Dehak-ro, Daejeon, Republic of Korea
| | - Aaron Z. Wagen
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Núria Setó-Salvia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Olga Kopach
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
- Neuroscience and Cell Biology Research Institute, City St George’s, University of London, Cranmer Terrace, London SW17 0RE, UK
| | - James R. Evans
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Margarida Rodrigues
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- UK Dementia Research Institute at The University of Cambridge, Cambridge CB2 0AH, UK
| | - Patricia Lopez-Garcia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Joanne Lachica
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Benjamin E. Clarke
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Jaijeet Singh
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Ali Ghareeb
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- Applied Biotechnology Lab, The Francis Crick Institute, London NW1 1AT, UK
| | - James Bayne
- Applied Biotechnology Lab, The Francis Crick Institute, London NW1 1AT, UK
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Melissa Grant-Peters
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Sonia Garcia-Ruiz
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Zhongbo Chen
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Samuel Rodriques
- Applied Biotechnology Lab, The Francis Crick Institute, London NW1 1AT, UK
- FutureHouse, 1405 Minnesota Street, San Francisco, CA 94107, USA
| | - Dilan Athauda
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Emil K. Gustavsson
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Sarah A. Gagliano Taliun
- Montréal Heart Institute, Montréal, QC, Canada
- Department of Medicine and Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Christina Toomey
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Regina H. Reynolds
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - George Young
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- MRC Laboratory of Medical Sciences, London W12 0HS, UK
| | - Stephanie Strohbuecker
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Thomas Warner
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Dmitri A. Rusakov
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Rickie Patani
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Clare Bryant
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK
| | - David A. Klenerman
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- UK Dementia Research Institute at The University of Cambridge, Cambridge CB2 0AH, UK
| | - Sonia Gandhi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Mina Ryten
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- UK Dementia Research Institute at The University of Cambridge, Cambridge CB2 0AH, UK
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| |
Collapse
|
63
|
Sirimaharaj N, Thiankhaw K, Chattipakorn N, Chattipakorn SC. Unveiling the Protective Roles of Melatonin on Glial Cells in the Battle Against Alzheimer's Disease-Insights from In Vivo and In Vitro Studies. Mol Neurobiol 2025:10.1007/s12035-025-04904-7. [PMID: 40208552 DOI: 10.1007/s12035-025-04904-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Alzheimer's disease (AD) is a chronic, progressive neurodegenerative disorder that predominantly affects the elderly. Characterized by amyloid-beta (Aβ) plaques and neurofibrillary tangles, AD leads to memory loss, cognitive decline, and severe behavioral changes. As the most common form of dementia, AD imposes a significant global health burden, highlighting the need for interventions that address underlying disease mechanisms rather than only symptomatic treatment. Glial cells, including microglia and astrocytes, play a crucial role in AD progression by mediating neuroinflammatory responses and modulating Aβ clearance and neuronal health. Dysfunction in these cells can exacerbate neuroinflammation and neuronal damage, making glial cells an important target for therapeutic intervention. This review synthesizes findings from in vivo and in vitro studies on melatonin's effects on glial cell dysfunction in AD, emphasizing the multi-mechanistic nature of its neuroprotective properties. Recent studies highlight melatonin's potential as a therapeutic agent that addresses AD-related mechanisms through its interactions with glial cells. Melatonin has demonstrated protective effects, including reducing oxidative stress, apoptosis, and inflammation, inhibiting Aβ fibrillogenesis, and modulating amyloid precursor proteins. Additionally, its influence on glial cell activity, through melatonin receptor pathways, suggests it can alleviate neuroinflammation, a key component of AD progression. The collective evidence points to melatonin's promise as a therapeutic tool with potential roles in both preventive and adjunctive treatments for AD. However, further research is necessary to establish its efficacy and safety in clinical settings.
Collapse
Affiliation(s)
- Nopdanai Sirimaharaj
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Kitti Thiankhaw
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
64
|
Zelic M, Blazier A, Pontarelli F, LaMorte M, Huang J, Tasdemir-Yilmaz OE, Ren Y, Ryan SK, Shapiro C, Morel C, Krishnaswami P, Levit M, Sood D, Chen Y, Gans J, Tang X, Hsiao-Nakamoto J, Huang F, Zhang B, Berry JD, Bangari DS, Gaglia G, Ofengeim D, Hammond TR. Single-cell transcriptomic and functional studies identify glial state changes and a role for inflammatory RIPK1 signaling in ALS pathogenesis. Immunity 2025; 58:961-979.e8. [PMID: 40132594 DOI: 10.1016/j.immuni.2025.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 10/31/2024] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by motor neuron loss. Microglia and astrocyte-driven neuroinflammation is prominent in ALS, but the cell state dynamics and pathways driving disease remain unclear. We performed single-nucleus RNA sequencing of ALS spinal cords and identified altered glial cell states, including increased expression of inflammatory and glial activation markers. Many of these signals converged on the inflammation and cell death regulator receptor-interacting protein kinase 1 (RIPK1) and the necroptotic cell death pathway. In superoxide dismutase 1 (SOD1)G93A mice, blocking RIPK1 kinase activity delayed symptom onset and motor impairment and modulated glial responses. We used human induced pluripotent stem cell (iPSC)-derived motor neuron, astrocyte, and microglia tri-cultures to identify potential biomarkers that are secreted upon RIPK1 activation in vitro and modulated by RIPK1 inhibition in the cerebrospinal fluid (CSF) of people with ALS. These data reveal ALS-enriched glial populations associated with inflammation and suggest a deleterious role for neuroinflammatory signaling in ALS pathogenesis.
Collapse
Affiliation(s)
- Matija Zelic
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA 02141, USA.
| | - Anna Blazier
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA 02141, USA
| | | | - Michael LaMorte
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA 02141, USA
| | - Jeremy Huang
- Sanofi, Precision Medicine and Computational Biology, Cambridge, MA 02141, USA
| | | | - Yi Ren
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA 02141, USA
| | - Sean K Ryan
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA 02141, USA
| | - Cynthia Shapiro
- Sanofi, Global Discovery Pathology and Multimodal Imaging, Cambridge, MA 02141, USA
| | - Caroline Morel
- Sanofi, Global Discovery Pathology and Multimodal Imaging, Cambridge, MA 02141, USA
| | | | - Mikhail Levit
- Sanofi, Precision Medicine and Computational Biology, Cambridge, MA 02141, USA
| | - Disha Sood
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA 02141, USA
| | - Yao Chen
- Sanofi, Precision Medicine and Computational Biology, Cambridge, MA 02141, USA
| | - Joseph Gans
- Sanofi, Precision Medicine and Computational Biology, Cambridge, MA 02141, USA
| | - Xinyan Tang
- Denali Therapeutics, Inc., South San Francisco, CA 94080, USA
| | | | - Fen Huang
- Denali Therapeutics, Inc., South San Francisco, CA 94080, USA
| | - Bailin Zhang
- Sanofi, Precision Medicine and Computational Biology, Cambridge, MA 02141, USA
| | - James D Berry
- Healey Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Dinesh S Bangari
- Sanofi, Global Discovery Pathology and Multimodal Imaging, Cambridge, MA 02141, USA
| | - Giorgio Gaglia
- Sanofi, Precision Medicine and Computational Biology, Cambridge, MA 02141, USA
| | | | | |
Collapse
|
65
|
Cai Y, Lin Z, Shen X, Li M, Xing L, Yang T, Chen G. Effect of microglial Pd1 on glial scar formation after spinal cord injury in mice. J Biol Chem 2025:108489. [PMID: 40209954 DOI: 10.1016/j.jbc.2025.108489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/27/2025] [Accepted: 04/02/2025] [Indexed: 04/12/2025] Open
Abstract
The crosstalk between microglia and astrocytes following spinal cord injury (SCI) greatly decides the prognosis. However, a comprehensive understanding of the molecular mechanisms by which microglia regulate astrocytic activity post-SCI is lacking. Programmed cell death protein 1 (Pdcd1, Pd1) plays a crucial role in modulating immune responses by exerting suppressive effects on microglia and peripheral immune cells within the central nervous system (CNS). Previous studies have shown the involvement of Pd1 in the pathogenesis of SCI; however, the role of microglial Pd1 in astrocytic activation and the following glial scar formation remains elusive. Here, we demonstrated that the pharmacological depletion of microglia using minocycline decreased the expression of TNF-α and IL-6 while concurrently increasing the expression of IL-10 following SCI, thereby facilitating motor function recovery in mice. We observed an increase in Pd1 expression in the injured spinal cord after SCI, with precise localization of Pd1 within microglia. Based on Pd1 knockout (KO) mice, we further revealed that Pd1 deficiency disrupted glial scar formation, leading to increased inflammation, impeded nerve regeneration, enlarged tissue damage, and compromised functional recovery following SCI. In vitro study showed that siRNA-mediated inhibition of Pd1 in microglia followed by lipopolysaccharide (LPS) treatment significantly inhibited astrocyte migration and upregulated the secretion of TNF-α and CXCL9 from microglia, indicating that microglial Pd1 regulates glial scar formation through modulating the inflammatory microenvironment. Our study gains a new mechanistic insight into how microglial Pd1 decides the fate of SCI and promotes microglial Pd1 as a promising therapeutic target for SCI.
Collapse
Affiliation(s)
- Yunyun Cai
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, 226001, China
| | - Zhihao Lin
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong, Jiangsu Province, 226001, China
| | - Xin Shen
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, 226001, China
| | - Ming Li
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, 226001, China
| | - Lingyan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong, Jiangsu Province, 226001, China
| | - Tuo Yang
- Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, China
| | - Gang Chen
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, 226001, China; Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong, Jiangsu Province, 226001, China; Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China.
| |
Collapse
|
66
|
Liu G, Li R, Gao J, Lin C, Li H, Peng Y, Wang H, Wang X. Mitigating Cannabidiol's Non-Selective Cytotoxicity via Subcellular Organelle Targeting: Exploring Mitochondrial Targeting Potential. Bioconjug Chem 2025. [PMID: 40198104 DOI: 10.1021/acs.bioconjchem.5c00012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Cannabidiol (CBD), a phytocannabinoid from Cannabis sativa, is renowned for its nonpsychoactive properties and therapeutic potential. However, its clinical application is limited by nonselective cytotoxicity, affecting microglia, oligodendrocytes, and other cells. To address this, subcellular organelle-targeting strategies were explored to minimize off-target effects and enhance CBD's therapeutic index. Three organelle-specific conjugates targeting mitochondria, endoplasmic reticulum, and lysosomes were synthesized. Among these, the mitochondria-targeting triphenylphosphonium (TPP)-modified CBD conjugates demonstrated reduced cytotoxicity and enhanced anti-inflammatory activity. Further optimization identified a four-carbon ether chain linker (CBD-TPP-C4) that increased antineuroinflammatory activity by 3-fold and reduced cytotoxicity by 1.6-fold, compared to unmodified CBD. CBD-TPP-C4 also elevated mitochondrial ATP levels in vitro, improved mitochondrial morphology and locomotor function in Caenorhabditis elegans, and potentiated morphine analgesia in mice. These findings highlight subcellular targeting as a promising strategy to enhance CBD's safety and efficacy, paving the way for improved therapeutic applications.
Collapse
Affiliation(s)
- Genglian Liu
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Ru Li
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China
| | - Jingwei Gao
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- Department of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Cong Lin
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Hongyuan Li
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Yinghua Peng
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China
| | - Hongshuang Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- Department of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
67
|
Fumagalli L, Nazlie Mohebiany A, Premereur J, Polanco Miquel P, Bijnens B, Van de Walle P, Fattorelli N, Mancuso R. Microglia heterogeneity, modeling and cell-state annotation in development and neurodegeneration. Nat Neurosci 2025:10.1038/s41593-025-01931-4. [PMID: 40195564 DOI: 10.1038/s41593-025-01931-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/20/2025] [Indexed: 04/09/2025]
Abstract
Within the CNS, microglia execute various functions associated with brain development, maintenance of homeostasis and elimination of pathogens and protein aggregates. This wide range of activities is closely associated with a plethora of cellular states, which may reciprocally influence or be influenced by their functional dynamics. Advancements in single-cell RNA sequencing have enabled a nuanced exploration of the intricate diversity of microglia, both in health and disease. Here, we review our current understanding of microglial transcriptional heterogeneity. We provide an overview of mouse and human microglial diversity encompassing aspects of development, neurodegeneration, sex and CNS regions. We offer an insight into state-of-the-art technologies and model systems that are poised to improve our understanding of microglial cell states and functions. We also provide suggestions and a tool to annotate microglial cell states on the basis of gene expression.
Collapse
Affiliation(s)
- Laura Fumagalli
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Alma Nazlie Mohebiany
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jessie Premereur
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Paula Polanco Miquel
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Baukje Bijnens
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Nicola Fattorelli
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Renzo Mancuso
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium.
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
68
|
Serrano GE, Aslam S, Walker JE, Piras IS, Huentelman MJ, Arce RA, Glass MJ, Intorcia AJ, Suszczewicz KE, Borja CI, Cline MP, Qiji SH, Lorenzini I, Beh ST, Mariner M, Krupp A, McHattie R, Shull A, Wermager ZR, Beach TG. Characterization of Isolated Human Astrocytes from Aging Brain. Int J Mol Sci 2025; 26:3416. [PMID: 40244314 PMCID: PMC11990013 DOI: 10.3390/ijms26073416] [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: 02/25/2025] [Revised: 03/27/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
Astrocytes have multiple crucial roles, including maintaining brain homeostasis and synaptic function, performing phagocytic clearance, and responding to injury and repair. It has been suggested that astrocyte performance is progressively impaired with aging, leading to imbalances in the brain's internal milieu that eventually impact neuronal function and lead to neurodegeneration. Until now, most evidence of astrocytic dysfunction in aging has come from experiments done with whole tissue homogenates, astrocytes collected by laser capture, or cell cultures derived from animal models or cell lines. In this study, we used postmortem-derived whole cells sorted with anti-GFAP antibodies to compare the unbiased, whole-transcriptomes of human astrocytes from control, older non-impaired individuals and subjects with different neurodegenerative diseases, such as Parkinson's disease (PD), Alzheimer's disease (ADD), and progressive supranuclear palsy (PSP). We found hundreds of dysregulated genes between disease and control astrocytes. In addition, we identified numerous genes shared between these common neurodegenerative disorders that are similarly dysregulated; in particular, UBC a gene for ubiquitin, which is a protein integral to cellular homeostasis and critically important in regulating function and outcomes of proteins under cellular stress, was upregulated in PSP, PD, and ADD when compared to control.
Collapse
Affiliation(s)
- Geidy E. Serrano
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (S.A.); (Z.R.W.)
| | - Sidra Aslam
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (S.A.); (Z.R.W.)
| | - Jessica E. Walker
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (S.A.); (Z.R.W.)
| | - Ignazio S. Piras
- Translational Genomics Research Institute, Phoenix, AZ 85004, USA; (I.S.P.); (M.J.H.)
| | - Matthew J. Huentelman
- Translational Genomics Research Institute, Phoenix, AZ 85004, USA; (I.S.P.); (M.J.H.)
| | - Richard A. Arce
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (S.A.); (Z.R.W.)
| | - Michael J. Glass
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (S.A.); (Z.R.W.)
| | - Anthony J. Intorcia
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (S.A.); (Z.R.W.)
| | | | - Claryssa I. Borja
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (S.A.); (Z.R.W.)
| | - Madison P. Cline
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (S.A.); (Z.R.W.)
| | - Sanaria H. Qiji
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (S.A.); (Z.R.W.)
| | - Ileana Lorenzini
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (S.A.); (Z.R.W.)
| | - Suet Theng Beh
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (S.A.); (Z.R.W.)
| | - Monica Mariner
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (S.A.); (Z.R.W.)
| | - Addison Krupp
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (S.A.); (Z.R.W.)
| | - Rylee McHattie
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (S.A.); (Z.R.W.)
| | - Anissa Shull
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (S.A.); (Z.R.W.)
| | - Zekiel R. Wermager
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (S.A.); (Z.R.W.)
| | - Thomas G. Beach
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (S.A.); (Z.R.W.)
| |
Collapse
|
69
|
Barones L, Weihs W, Schratter A, Janata A, Kodajova P, Bergmeister H, Kenner L, Holzer M, Behringer W, Högler S. Cold aortic flush after ventricular fibrillation cardiac arrest reduces inflammatory reaction but not neuronal loss in the pig cerebral cortex. Sci Rep 2025; 15:11659. [PMID: 40185805 PMCID: PMC11971268 DOI: 10.1038/s41598-025-95611-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
This study aims to retrospectively compare two resuscitation methods (extracorporeal cardiopulmonary resuscitation (ECPR) vs. emergency preservation and resuscitation (EPR)) by pathohistologically assessing pig brains in a ventricular fibrillation cardiac arrest (VFCA) model. In prospective studies from 2004 to 2006, swine underwent VFCA for 13 (n = 6), 15 (n = 14) or 17 (n = 6) minutes with ECPR (ECPR13, ECPR15 and ECPR17). Another 15 min VFCA group (n = 8) was resuscitated with EPR and chest compressions (EPR15 + CC). Brains of animals surviving for nine days (ECPR13 n = 4, ECPR15 n = 2, ECPR17 n = 1, EPR15 + CC n = 7) were harvested. Eight different brain regions were analyzed with the image analysis software QuPath using HE-staining, GFAP- and Iba1-immunohistochemistry. Only ECPR13 and EPR15 + CC animals were included in statistical analysis, due to low survival rates in the other groups. All VFCA samples showed significantly fewer viable neurons compared to shams, but no significant differences between ECPR13 and EPR15 + CC animals were observed. ECPR13 animals showed significantly more glial activation in all cerebral cortex regions compared to shams and in occipital, temporal and parietal cortex compared to EPR15 + CC. In conclusion, EPR + CC resulted in a significantly reduced inflammatory reaction in cerebral cortex compared to ECPR but did not influence the extent of neuronal death after VFCA.
Collapse
Affiliation(s)
- Lisa Barones
- Laboratory Animal Pathology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Wolfgang Weihs
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Andreas Janata
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Petra Kodajova
- Laboratory Animal Pathology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Helga Bergmeister
- Center for Biomedical Research and Translational Surgery and Ludwig Boltzmann Institute for Cardiovascular Research, Medical University Vienna, Vienna, Austria
| | - Lukas Kenner
- Laboratory Animal Pathology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- Department of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
| | - Michael Holzer
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Wilhelm Behringer
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Sandra Högler
- Laboratory Animal Pathology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria.
| |
Collapse
|
70
|
Huang L, Li Q, Wu J, He Y, Huang J, Xie S, Yang C, Ruan Q, Zhou Z, Deng M. Galangin reduces MPTP-induced dopamine neuron injury via the autophagy dependent-PI3K/AKT pathway. Front Aging Neurosci 2025; 17:1568002. [PMID: 40256391 PMCID: PMC12006098 DOI: 10.3389/fnagi.2025.1568002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/17/2025] [Indexed: 04/22/2025] Open
Abstract
Introduction Research has confirmed that Galangin can attenuate autophagy and protect dopaminergic neurons. This study aims to clarify whether Galangin attenuates dopaminergic neuron injury by regulating the PI3K/AKT pathway in Parkinson's disease (PD) model mice. Methods The study explores the mitigating effects of Galangin on PD processes by administering 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to induce the condition. Techniques including network analysis, transcriptomic analysis, rotarod test, enzyme-linked immunosorbent assay (ELISA), qRT-PCR, western blotting, immunohistochemistry, immunofluorescence, and hematoxylin-eosin (HE) were employed to unveil the molecular changes induced by Galangin. Results The network pharmacological analysis showed 301 targets related to Galangin, and 2,858 genes related to PD. Galangin treatment can improve the motor coordination of PD model mice, reduce damage to neurons in the brain, improve the antioxidant capacity and reduce the inflammatory damage of brain tissue. Additionally, Galangin suppressed mRNA expression of PD markers (IL-1β, TNF-α, IL-6, SRC and PTGS2), elevated protein levels of GSH-Px, SOD, P-PI3K, P-CREB, P-AKT, TH, BDNF and P62, while decreasing α-syn, SRC, MDA, Beclin-1 and LC3B expression. Moreover, the expression of significantly different genes in the Galangin-treated group and model group analyzed by transcriptomics was basically consistent with the qRT-PCR verification results. Conclusion Galangin supresses Beclin-1-dependent autophagy and upregulates the PI3K/AKT signaling pathway to attenuate the neuroinflammatory injury and improve motor coordination ability in PD mice induced by MPTP.
Collapse
Affiliation(s)
- Liping Huang
- School of Chemistry and Chemical Engineering, Western Guangdong Characteristic Biomedical Engineering Technology Research Center, Lingnan Normal University, Zhanjiang, China
- Mangrove Institute, Lingnan Normal University, Zhanjiang, China
| | - Qiaofeng Li
- School of Chemistry and Chemical Engineering, Western Guangdong Characteristic Biomedical Engineering Technology Research Center, Lingnan Normal University, Zhanjiang, China
| | - Jingyi Wu
- School of Chemistry and Chemical Engineering, Western Guangdong Characteristic Biomedical Engineering Technology Research Center, Lingnan Normal University, Zhanjiang, China
| | - Yingying He
- School of Chemistry and Chemical Engineering, Western Guangdong Characteristic Biomedical Engineering Technology Research Center, Lingnan Normal University, Zhanjiang, China
| | - Junwei Huang
- School of Chemistry and Chemical Engineering, Western Guangdong Characteristic Biomedical Engineering Technology Research Center, Lingnan Normal University, Zhanjiang, China
| | - Sipeng Xie
- School of Chemistry and Chemical Engineering, Western Guangdong Characteristic Biomedical Engineering Technology Research Center, Lingnan Normal University, Zhanjiang, China
| | - Canfeng Yang
- School of Chemistry and Chemical Engineering, Western Guangdong Characteristic Biomedical Engineering Technology Research Center, Lingnan Normal University, Zhanjiang, China
| | - Qingling Ruan
- School of Chemistry and Chemical Engineering, Western Guangdong Characteristic Biomedical Engineering Technology Research Center, Lingnan Normal University, Zhanjiang, China
| | - Zhongliu Zhou
- School of Chemistry and Chemical Engineering, Western Guangdong Characteristic Biomedical Engineering Technology Research Center, Lingnan Normal University, Zhanjiang, China
| | - Minzhen Deng
- State Key Laboratory of Traditional Chinese Medicine Syndrome/ Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou, China
| |
Collapse
|
71
|
Sim M, Hong S, Jung MH, Choi EY, Hwang GS, Shin DM, Kim CS. Gut microbiota links vitamin C supplementation to enhanced mental vitality in healthy young adults with suboptimal vitamin C status: A randomized, double-blind, placebo-controlled trial. Brain Behav Immun 2025; 128:179-191. [PMID: 40187667 DOI: 10.1016/j.bbi.2025.03.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/07/2025] [Accepted: 03/25/2025] [Indexed: 04/07/2025] Open
Abstract
The intricate relationship between nutrition, gut microbiome, and mental health has gained increasing attention. We aimed to determine how vitamin C supplementation improves mental vitality through the gut microbiome and associated neurological and immunological changes. We used 16S rRNA sequencing to analyze gut microbiota profiles of participants from our previous trial, in which healthy young adults (20-39 years) with inadequate serum vitamin C levels (< 50 μM) received 500 mg vitamin C or a placebo twice daily for 4 weeks (vitamin C, n = 21; placebo, n = 19). We examined whether changes in gut microbiota correlated with previously determined mental vitality indices, including Stroop test performance, work engagement, and serum brain-derived neurotrophic factor (BDNF) levels. Serum concentrations of microbial-derived molecules, cytokines, and neurotransmitters were analyzed using enzyme-linked immunosorbent assay, electrochemiluminescence-based immunoassay, or ultra-high-performance liquid chromatography-mass spectrometry. Monocyte subpopulations in peripheral blood were quantified using fluorescence-activated cell sorting analysis. Vitamin C supplementation increased the relative abundance of Bacillaceae and Anaerotruncus, while decreasing Desulfovibrio, with the Desulfovibrio reduction correlating with Stroop test performance. Moreover, participants showing a substantial Desulfovibrio reduction ("responders") demonstrated greater BDNF increases and stronger correlations between serum L-DOPA levels and work engagement scores than did non-responders. In addition, vitamin C supplementation suppressed inflammatory responses with concurrent reduction in serum lipopolysaccharide levels, and responders showed greater decreases in IL-10 levels and classical monocyte frequencies than non-responders. In conclusion, vitamin C supplementation modulates gut microbiota composition, particularly by reducing Desulfovibrio abundance, with the extent of reduction correlating with mental vitality improvements and decreased inflammation. This study provides insights into vitamin C supplementation as a critical dietary intervention, as it may modulate mental health through its influence on the gut-brain-immune axis.
Collapse
Affiliation(s)
- Minju Sim
- Department of Food and Nutrition, Seoul National University, Seoul 08826, Republic of Korea
| | - Sehwa Hong
- Department of Food and Nutrition, Seoul National University, Seoul 08826, Republic of Korea
| | - Min Ho Jung
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Eun Young Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul 03759, Republic of Korea; College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Dong-Mi Shin
- Department of Food and Nutrition, Seoul National University, Seoul 08826, Republic of Korea
| | - Chong-Su Kim
- Department of Food and Nutrition, Seowon University, Cheongju 28674, Republic of Korea.
| |
Collapse
|
72
|
Vicario R, Fragkogianni S, Pokrovskii M, Meyer C, Lopez-Rodrigo E, Hu Y, Ogishi M, Alberdi A, Baako A, Ay O, Plu I, Sazdovitch V, Heritier S, Cohen-Aubart F, Shor N, Miyara M, Nguyen-Khac F, Viale A, Idbaih A, Amoura Z, Rosenblum MK, Zhang H, Karnoub ER, Sashittal P, Jakatdar A, Iacobuzio-Donahue CA, Abdel-Wahab O, Tabar V, Socci ND, Elemento O, Diamond EL, Boisson B, Casanova JL, Seilhean D, Haroche J, Donadieu J, Geissmann F. Role of clonal inflammatory microglia in histiocytosis-associated neurodegeneration. Neuron 2025; 113:1065-1081.e13. [PMID: 40081365 DOI: 10.1016/j.neuron.2025.02.007] [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: 07/12/2023] [Revised: 10/28/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025]
Abstract
Langerhans cell histiocytosis (LCH) and Erdheim-Chester disease (ECD) are clonal myeloid disorders associated with mitogen-activated protein (MAP)-kinase-activating mutations and an increased risk of neurodegeneration. We found microglial mutant clones in LCH and ECD patients, whether or not they presented with clinical symptoms of neurodegeneration, associated with microgliosis, astrocytosis, and neuronal loss, predominantly in the rhombencephalon gray nuclei. Neurological symptoms were associated with PU.1+ clone size (p = 0.0003) in patients with the longest evolution of the disease, indicating a phase of subclinical incipient neurodegeneration. Genetic barcoding analysis suggests that clones may originate from definitive or yolk sac hematopoiesis, depending on the patients. In a mouse model, disease topography was attributable to a local clonal proliferative advantage, and microglia depletion by a CSF1R-inhibitor limited neuronal loss and improved survival. These studies characterize a neurodegenerative disease associated with clonal proliferation of inflammatory microglia. The long preclinical stage represents a therapeutic window before irreversible neuronal depletion.
Collapse
Affiliation(s)
- Rocio Vicario
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stamatina Fragkogianni
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Maria Pokrovskii
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Carina Meyer
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Estibaliz Lopez-Rodrigo
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yang Hu
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell, New York, NY 10021, USA
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Araitz Alberdi
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ann Baako
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Oyku Ay
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Isabelle Plu
- Department of Neuropathology, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | - Véronique Sazdovitch
- Department of Neuropathology, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | - Sebastien Heritier
- French Langerhans cell histiocytosis registry, Department of Pediatric Hematology and Oncology, Trousseau Hospital, AP-HP, Paris, France
| | - Fleur Cohen-Aubart
- Department of Internal Medicine & Institut E3M, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | - Natalia Shor
- Department of Neuroradiology, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | - Makoto Miyara
- Center for Immunology and Infectious Diseases (CIMI-PARIS), Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | - Florence Nguyen-Khac
- Department of Hematology, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | - Agnes Viale
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, MSKCC, New York, NY 10065, USA
| | - Ahmed Idbaih
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié-Salpêtrière - Charles Foix, Service de Neurologie 2-Mazarin, 75013 Paris, France
| | - Zahir Amoura
- Department of Neuroradiology, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | | | - Haochen Zhang
- Human Oncology and Pathogenesis Program, MSKCC, New York, NY, USA
| | | | - Palash Sashittal
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Akhil Jakatdar
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Christine A Iacobuzio-Donahue
- Department of Pathology, MSKCC, New York, NY 10065, USA; Human Oncology and Pathogenesis Program, MSKCC, New York, NY, USA
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, MSKCC, New York, NY, USA
| | - Viviane Tabar
- Department of Neurosurgery, and Center for Stem Cell Biology, MSKCC, New York, NY, USA; Department of Neurology, MSKCC, New York, NY 10065, USA
| | - Nicholas D Socci
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, MSKCC, New York, NY 10065, USA
| | - Olivier Elemento
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell, New York, NY 10021, USA
| | - Eli L Diamond
- Department of Neurosurgery, and Center for Stem Cell Biology, MSKCC, New York, NY, USA
| | - Bertrand Boisson
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Danielle Seilhean
- Department of Neuropathology, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | - Julien Haroche
- Department of Internal Medicine & Institut E3M, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France.
| | - Jean Donadieu
- French Langerhans cell histiocytosis registry, Department of Pediatric Hematology and Oncology, Trousseau Hospital, AP-HP, Paris, France
| | - Frederic Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
73
|
Yu Y, Wang T, Li Q, Zhao H, Li B, Lei D, Dong F, Xiao Y, Wang S, Ji Y. DL-3-n-butylphthalide inhibits astrocyte activation in the cortical penumbra of ischemia-reperfusion model rats via AKT signaling. Brain Res Bull 2025; 225:111332. [PMID: 40185418 DOI: 10.1016/j.brainresbull.2025.111332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/23/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
Ischemic stroke triggers rapid activation of astrocytes, which contributes to tissue damage. Dl-3-n-butylphthalide (NBP), an independently developed compound in China for the treatment of ischemic stroke, has unclear molecular mechanisms. In this study, we established a Sprague-Dawley rat model of middle cerebral artery occlusion (MCAO) by occluding the middle cerebral artery for 1.5 h followed by reperfusion for 72 h. We assessed neurological scores, infarct volume, neuronal injury, and the expression levels of GFAP, C3, S100A10, GLT-1, p-AKT/AKT, and p-mTOR/mTOR, as well as immunofluorescence double staining of C3/S100A10 with GFAP and GLT-1 respectively. NBP significantly improved neurological function in MCAO rats, reduced infarct area, alleviated neuronal injury, inhibited A1 astrocyte polarization, promoted A2 astrocyte polarization, and upregulated GLT-1 expression. However, the AKT inhibitor (TCN) weakened NBP's regulatory effects on astrocytes and GLT-1. Finally, immunofluorescence experiments showed that GLT-1 colocalized more effectively with A2 astrocytes than with A1 astrocytes. We demonstrated that NBP reduces astrocyte activation and upregulates GLT-1 expression via the AKT/mTOR pathway, providing new insights into therapeutic strategies for ischemic stroke and valuable clues for drug design.
Collapse
Affiliation(s)
- Yiwen Yu
- Department of Neurology, Nanchong Central Hospital Affiliated to North Sichuan Medical College, Nanchong, Sichuan, China
| | - Tinghong Wang
- Department of Forensic Pathology, School of Basic Medical Science and Forensic Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Qiuling Li
- Department of Neurology, Nanchong Central Hospital Affiliated to North Sichuan Medical College, Nanchong, Sichuan, China
| | - Hao Zhao
- Department of Neurology, Nanchong Central Hospital Affiliated to North Sichuan Medical College, Nanchong, Sichuan, China
| | - Biao Li
- Department of Neurology, Nanchong Central Hospital Affiliated to North Sichuan Medical College, Nanchong, Sichuan, China
| | - Dong Lei
- Department of Neurology, Nanchong Central Hospital Affiliated to North Sichuan Medical College, Nanchong, Sichuan, China
| | - Fei Dong
- Department of Neurology, Nanchong Central Hospital Affiliated to North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yu Xiao
- Department of Neurology, Nanchong Central Hospital Affiliated to North Sichuan Medical College, Nanchong, Sichuan, China
| | - Shan Wang
- Department of Neurology, Nanchong Central Hospital Affiliated to North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yifei Ji
- Department of Neurology, Nanchong Central Hospital Affiliated to North Sichuan Medical College, Nanchong, Sichuan, China.
| |
Collapse
|
74
|
Lawrence AB, Brown SM, Bradford BM, Mabbott NA, Bombail V, Rutherford KMD. Non-neuronal brain biology and its relevance to animal welfare. Neurosci Biobehav Rev 2025; 173:106136. [PMID: 40185375 DOI: 10.1016/j.neubiorev.2025.106136] [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: 07/10/2024] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Non-neuronal cells constitute a significant portion of brain tissue and are seen as having key roles in brain homeostasis and responses to challenges. This review illustrates how non-neuronal biology can bring new perspectives to animal welfare through understanding mechanisms that determine welfare outcomes and highlighting interventions to improve welfare. Most obvious in this respect is the largely unrecognised relevance of neuroinflammation to animal welfare which is increasingly found to have roles in determining how animals respond to challenges. We start by introducing non-neuronal cells and review their involvement in affective states and cognition often seen as core psychological elements of animal welfare. We find that the evidence for a causal involvement of glia in cognition is currently more advanced than the corresponding evidence for affective states. We propose that translational research on affective disorders could usefully apply welfare science derived approaches for assessing affective states. Using evidence from translational research, we illustrate the involvement of non-neuronal cells and neuroinflammatory processes as mechanisms modulating resilience to welfare challenges including disease, pain, and social stress. We review research on impoverished environments and environmental enrichment which suggests that environmental conditions which improve animal welfare also improve resilience to challenges through balancing pro- and anti-inflammatory non-neuronal processes. We speculate that non-neuronal biology has relevance to animal welfare beyond neuro-inflammation including facilitating positive affective states. We acknowledge the relevance of neuronal biology to animal welfare whilst proposing that non-neuronal biology provides additional and relevant insights to improve animals' lives.
Collapse
Affiliation(s)
- Alistair B Lawrence
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK; Scotland's Rural College (SRUC), Edinburgh EH9 3JG, UK.
| | - Sarah M Brown
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Barry M Bradford
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Neil A Mabbott
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | | | | |
Collapse
|
75
|
Campos J, Palha AT, Fernandes LS, Cibrão JR, Pinho TS, Serra SC, Silva NA, Michael-Titus AT, Salgado AJ. Modeling Spinal Cord Injury in a Dish with Hyperosmotic Stress: Population-Specific Effects and the Modulatory Role of Mesenchymal Stromal Cell Secretome. Int J Mol Sci 2025; 26:3298. [PMID: 40244122 PMCID: PMC11989751 DOI: 10.3390/ijms26073298] [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: 01/17/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Innovations in spinal cord injury (SCI) models are crucial for developing effective therapies. This study introduces a novel in vitro SCI model using cultures of primary mixed spinal cord cells from rat pups, featuring key spinal cord cell types. This model offers distinct advantages in terms of feasibility, reproducibility, and cost-effectiveness, requiring only basic cell culture equipment. Following hyperosmotic stress via sorbitol treatment, the model recapitulated SCI pathophysiological hallmarks, with a 65% reduction in cell viability and gradual cell death over 48 h, making it ideal for evaluating neuroprotective agents. Notably, the human adipose tissue stem cell (hASC) secretome provided significant protection: it preserved metabolic viability, reduced β amyloid precursor protein (β-APP) expression in surviving neurons, and modulated the shift in the astrocytic morphotype. A transcriptomic profile of the effect of the hASC secretome treatment showed significant functional enrichments related to cell proliferation and cycle progression pathways. In addition to supporting the use of the hASC secretome as a therapy for SCI, this study is the first to use sorbitol as a hyperosmolar stressor to recapitulate key aspects of SCI pathophysiology. Thereby, this model can be used as a promising platform for evaluating therapeutic agents targeting neuroprotection and neuroregeneration, offering outputs related to cell death, neuronal stress, and protection, as well as induction of glial reactivity.
Collapse
Affiliation(s)
- Jonas Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.C.); (A.T.P.); (L.S.F.); (J.R.C.); (T.S.P.); (S.C.S.); (N.A.S.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimaraes, Portugal
| | - Ana T. Palha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.C.); (A.T.P.); (L.S.F.); (J.R.C.); (T.S.P.); (S.C.S.); (N.A.S.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimaraes, Portugal
| | - Luís S. Fernandes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.C.); (A.T.P.); (L.S.F.); (J.R.C.); (T.S.P.); (S.C.S.); (N.A.S.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimaraes, Portugal
| | - Jorge R. Cibrão
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.C.); (A.T.P.); (L.S.F.); (J.R.C.); (T.S.P.); (S.C.S.); (N.A.S.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimaraes, Portugal
| | - Tiffany S. Pinho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.C.); (A.T.P.); (L.S.F.); (J.R.C.); (T.S.P.); (S.C.S.); (N.A.S.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimaraes, Portugal
| | - Sofia C. Serra
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.C.); (A.T.P.); (L.S.F.); (J.R.C.); (T.S.P.); (S.C.S.); (N.A.S.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimaraes, Portugal
| | - Nuno A. Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.C.); (A.T.P.); (L.S.F.); (J.R.C.); (T.S.P.); (S.C.S.); (N.A.S.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimaraes, Portugal
| | - Adina T. Michael-Titus
- Centre for Neuroscience, Surgery and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK;
| | - António J. Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.C.); (A.T.P.); (L.S.F.); (J.R.C.); (T.S.P.); (S.C.S.); (N.A.S.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimaraes, Portugal
| |
Collapse
|
76
|
König J, Blusch A, Fatoba O, Gold R, Saft C, Ellrichmann-Wilms G. Examination of Anti-Inflammatory Effects After Propionate Supplementation in the R6/2 Mouse Model of Huntington's Disease. Int J Mol Sci 2025; 26:3318. [PMID: 40244185 PMCID: PMC11989372 DOI: 10.3390/ijms26073318] [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: 01/27/2025] [Revised: 03/23/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Huntington's disease is a progressive, untreatable neurodegenerative disorder caused by a mutation in the Huntingtin gene. Next to neurodegeneration, altered immune activation is involved in disease progression. Since central nervous system inflammation and dysfunction of immune cells are recognized as driving characteristics, immunomodulation might represent an additional therapeutic strategy. Short-chain fatty acids were known to have immunomodulatory effects in neuroinflammatory diseases, such as multiple sclerosis. In this study, R6/2 mice were treated daily with 150 mM propionate. Survival range, body weight, and motor abilities were monitored. In striatal and cortical samples, neuronal survival was analyzed by immunofluorescence staining of NeuN-positive cells and expression levels of BDNF mRNA by real-time polymerase chain reaction. As inflammatory marker TNFα mRNA and IL-6 mRNA were quantified by rtPCR, iNOS-expressing cells were counted in immunologically stained brain slides. Microglial activation was evaluated by immunofluorescent staining of IBA1-positive cells and total IBA1 protein by Western Blot, in addition, SPI1 mRNA expression was quantified by rtPCR. Except for clasping behavior, propionate treatment did neither improve the clinical course nor mediated neuronal protection in R6/2 mice. Yet there was a mild anti-inflammatory effect in the CNS, with (i) reduction in SPI1-mRNA levels, (ii) reduced iNOS positive cells in the motor cortex, and (iii) normalized TNFα-mRNA in the motor cortex of propionate-treated R6/2 mice. Thus, Short-chain fatty acids, as an environmental factor in the diet, may slightly alleviate symptoms by down-regulating inflammatory factors in the central nervous system. However, they cannot prevent clinical disease progression or neuronal loss.
Collapse
Affiliation(s)
- Jennifer König
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
- Department of Physiology and Pathophysiology, Center of Biomedical Education and Research (ZBAF), Faculty of Health, School of Medicine, Witten/Herdecke University, 58453 Witten, Germany
| | - Alina Blusch
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
- Brain Disease Biomarker Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, BMC A10, 221 84 Lund, Sweden
| | - Oluwaseun Fatoba
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
| | - Ralf Gold
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Carsten Saft
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Gisa Ellrichmann-Wilms
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
- Faculty of Health, School of Medicine, Chair of Neurology II, Witten/Herdecke University, 58448 Witten, Germany
| |
Collapse
|
77
|
Molitor TP, Hayashi G, Lin MY, Dunn CJ, Peterson NG, Poston RG, Kurnellas MP, Traver DA, Patel S, Akgungor Z, Leonardi V, Lewis C, Segales JS, Bennett DS, Truong AP, Dani M, Naphade S, Wong JK, McDermott AE, Kovalev SM, Ciaccio GL, Sadiq SA, Pei Z, Wood S, Rassoulpour A. Central TYK2 inhibition identifies TYK2 as a key neuroimmune modulator. Proc Natl Acad Sci U S A 2025; 122:e2422172122. [PMID: 40127268 PMCID: PMC12002270 DOI: 10.1073/pnas.2422172122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/03/2025] [Indexed: 03/26/2025] Open
Abstract
GWAS have identified tyrosine kinase 2 (TYK2) variants in multiple inflammatory disorders, specifically a protective hypomorphic TYK2 allele (P1104A) in multiple sclerosis (MS). Impaired TYK2 signaling within the central nervous system (CNS) may impart the protective effects of TYK2 P1104A allele in MS. We deployed brain-penetrant TYK2 inhibitors (cTYK2i) alongside the peripherally restricted TYK2 inhibitor (pTYK2i; BMS-986165) to untangle the contributions of central TYK2 inhibition in diverse models of neuroinflammation. While pTYK2i had little impact, cTYK2i reduced clinical score, lymphoid cell infiltration, and cytokines/chemokines in experimental autoimmune encephalomyelitis (EAE). Microglial activation was attenuated in cTYK2i-treated EAE spinal cords and circulating neurofilament light (NfL) was reduced in plasma and cerebral spinal fluid (CSF). Additionally, cTYK2i was protective in an antibody-mediated mouse model of primary progressive MS (PPMS). Finally, we demonstrate TYK2 inhibition has a robust impact on a unique subset of activated astrocytes termed Interferon-Responsive-Reactive-Astrocytes (IRRA). The data presented herein identify a key role for CNS TYK2 signaling in regulating neuroinflammation and solidify TYK2 as a potential therapeutic target for MS.
Collapse
Affiliation(s)
- Tyler P. Molitor
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Genki Hayashi
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Mei-Yao Lin
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Carissa J. Dunn
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | | | - Robert G. Poston
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | | | - David A. Traver
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Seona Patel
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Zeynep Akgungor
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | | | - Colizel Lewis
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | | | - Dylan S. Bennett
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Anh P. Truong
- Department of Chemistry, Neuron23, Inc., South San Francisco, CA94080
| | - Manjari Dani
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Swati Naphade
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | - Jamie K. Wong
- Tisch MS Research Center of New York, New York, NY10019
| | | | | | | | - Saud A. Sadiq
- Tisch MS Research Center of New York, New York, NY10019
| | - Zhonghua Pei
- Department of Chemistry, Neuron23, Inc., South San Francisco, CA94080
| | - Stephen Wood
- Department of Biology, Neuron23, Inc., South San Francisco, CA94080
| | | |
Collapse
|
78
|
Ziar R, Tesar PJ, Clayton BLL. Astrocyte and oligodendrocyte pathology in Alzheimer's disease. Neurotherapeutics 2025; 22:e00540. [PMID: 39939240 DOI: 10.1016/j.neurot.2025.e00540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 01/10/2025] [Accepted: 01/24/2025] [Indexed: 02/14/2025] Open
Abstract
Astrocytes and oligodendrocytes, once considered passive support cells, are now recognized as active participants in the pathogenesis of Alzheimer's disease. Emerging evidence highlights the critical role that these glial cells play in the pathological features of Alzheimer's, including neuroinflammation, excitotoxicity, synaptic dysfunction, and myelin degeneration, which contribute to neurodegeneration and cognitive decline. Here, we review the current understanding of astrocyte and oligodendrocyte pathology in Alzheimer's disease and highlight research that supports the therapeutic potential of modulating astrocyte and oligodendrocyte functions to treat Alzheimer's disease.
Collapse
Affiliation(s)
- Rania Ziar
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Paul J Tesar
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Benjamin L L Clayton
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
79
|
Xu W, Huang Y, Zhou R. NLRP3 inflammasome in neuroinflammation and central nervous system diseases. Cell Mol Immunol 2025; 22:341-355. [PMID: 40075143 PMCID: PMC11955557 DOI: 10.1038/s41423-025-01275-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Neuroinflammation plays an important role in the pathogenesis of various central nervous system (CNS) diseases. The NLRP3 inflammasome is an important intracellular multiprotein complex composed of the innate immune receptor NLRP3, the adaptor protein ASC, and the protease caspase-1. The activation of the NLRP3 inflammasome can induce pyroptosis and the release of the proinflammatory cytokines IL-1β and IL-18, thus playing a central role in immune and inflammatory responses. Recent studies have revealed that the NLRP3 inflammasome is activated in the brain to induce neuroinflammation, leading to further neuronal damage and functional impairment, and contributes to the pathological process of various neurological diseases, such as multiple sclerosis, Parkinson's disease, Alzheimer's disease, and stroke. In this review, we summarize the important role of the NLRP3 inflammasome in the pathogenesis of neuroinflammation and the pathological course of CNS diseases and discuss potential approaches to target the NLRP3 inflammasome for the treatment of CNS diseases.
Collapse
Grants
- 81821001, 82130107, 82330052, 82202038, U20A20359 National Natural Science Foundation of China (National Science Foundation of China)
- National Key research and development program of China (grant number (2020YFA0509101), The Strategic Priority Research Program of the Chinese Academy of Sciences (XDB0940000),
- MEXT | JST | Strategic Promotion of Innovative R and D (Strategic Promotion of Innovative R&D)
- the CAS Project for Young Scientists in Basic Research (YSBR-074) and the Fundamental Research Funds for the Central Universities, the outstanding Youth Project of Anhui Provincial Natural Science Foundation (2408085Y049), the Research Start-up Funding of the Institute of Health and Medicine, Hefei Comprehensive National Science Center (2024KYQD004), the Natural Science Foundation of Jiangsu Province (BK20221085),
- The key project of Anhui Provincial Department of Education Fund (2024AH052060).
Collapse
Affiliation(s)
- Wen Xu
- Neurology Department, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, P. R. China
| | - Yi Huang
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230601, China.
| | - Rongbin Zhou
- National Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, China.
- Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
| |
Collapse
|
80
|
Cieri MB, Ramos AJ. Astrocytes, reactive astrogliosis, and glial scar formation in traumatic brain injury. Neural Regen Res 2025; 20:973-989. [PMID: 38989932 PMCID: PMC11438322 DOI: 10.4103/nrr.nrr-d-23-02091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 04/14/2024] [Indexed: 07/12/2024] Open
Abstract
Traumatic brain injury is a global health crisis, causing significant death and disability worldwide. Neuroinflammation that follows traumatic brain injury has serious consequences for neuronal survival and cognitive impairments, with astrocytes involved in this response. Following traumatic brain injury, astrocytes rapidly become reactive, and astrogliosis propagates from the injury core to distant brain regions. Homeostatic astroglial proteins are downregulated near the traumatic brain injury core, while pro-inflammatory astroglial genes are overexpressed. This altered gene expression is considered a pathological remodeling of astrocytes that produces serious consequences for neuronal survival and cognitive recovery. In addition, glial scar formed by reactive astrocytes is initially necessary to limit immune cell infiltration, but in the long term impedes axonal reconnection and functional recovery. Current therapeutic strategies for traumatic brain injury are focused on preventing acute complications. Statins, cannabinoids, progesterone, beta-blockers, and cerebrolysin demonstrate neuroprotective benefits but most of them have not been studied in the context of astrocytes. In this review, we discuss the cell signaling pathways activated in reactive astrocytes following traumatic brain injury and we discuss some of the potential new strategies aimed to modulate astroglial responses in traumatic brain injury, especially using cell-targeted strategies with miRNAs or lncRNA, viral vectors, and repurposed drugs.
Collapse
Affiliation(s)
- María Belén Cieri
- Laboratorio de Neuropatología Molecular, IBCN UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | |
Collapse
|
81
|
Vanherle S, Loix M, Miron VE, Hendriks JJA, Bogie JFJ. Lipid metabolism, remodelling and intercellular transfer in the CNS. Nat Rev Neurosci 2025; 26:214-231. [PMID: 39972160 DOI: 10.1038/s41583-025-00908-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2025] [Indexed: 02/21/2025]
Abstract
Lipid metabolism encompasses the catabolism and anabolism of lipids, and is fundamental for the maintenance of cellular homeostasis, particularly within the lipid-rich CNS. Increasing evidence further underscores the importance of lipid remodelling and transfer within and between glial cells and neurons as key orchestrators of CNS lipid homeostasis. In this Review, we summarize and discuss the complex landscape of processes involved in lipid metabolism, remodelling and intercellular transfer in the CNS. Highlighted are key pathways, including those mediating lipid (and lipid droplet) biogenesis and breakdown, lipid oxidation and phospholipid metabolism, as well as cell-cell lipid transfer mediated via lipoproteins, extracellular vesicles and tunnelling nanotubes. We further explore how the dysregulation of these pathways contributes to the onset and progression of neurodegenerative diseases, and examine the homeostatic and pathogenic impacts of environment, diet and lifestyle on CNS lipid metabolism.
Collapse
Affiliation(s)
- Sam Vanherle
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Centre, Hasselt University, Hasselt, Belgium
| | - Melanie Loix
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Centre, Hasselt University, Hasselt, Belgium
| | - Veronique E Miron
- Keenan Research Centre for Biomedical Science and Barlo Multiple Sclerosis Centre, St Michael's Hospital, Toronto, Ontario, Canada
- Department of Immunology, The University of Toronto, Toronto, Ontario, Canada
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
| | - Jerome J A Hendriks
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Centre, Hasselt University, Hasselt, Belgium
| | - Jeroen F J Bogie
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.
- University MS Centre, Hasselt University, Hasselt, Belgium.
| |
Collapse
|
82
|
Liu Y, Cai X, Shi B, Mo Y, Zhang J, Luo W, Yu B, Li X. Mechanisms and Therapeutic Prospects of Microglia-Astrocyte Interactions in Neuropathic Pain Following Spinal Cord Injury. Mol Neurobiol 2025; 62:4654-4676. [PMID: 39470872 DOI: 10.1007/s12035-024-04562-1] [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: 05/24/2024] [Accepted: 10/16/2024] [Indexed: 11/01/2024]
Abstract
Neuropathic pain is a prevalent and debilitating condition experienced by the majority of individuals with spinal cord injury (SCI). The complex pathophysiology of neuropathic pain, involving continuous activation of microglia and astrocytes, reactive gliosis, and altered neuronal plasticity, poses significant challenges for effective treatment. This review focuses on the pivotal roles of microglia and astrocytes, the two major glial cell types in the central nervous system, in the development and maintenance of neuropathic pain after SCI. We highlight the extensive bidirectional interactions between these cells, mediated by the release of inflammatory mediators, neurotransmitters, and neurotrophic factors, which contribute to the amplification of pain signaling. Understanding the microglia-astrocyte crosstalk and its impact on neuronal function is crucial for developing novel therapeutic strategies targeting neuropathic pain. In addition, this review discusses the fundamental biology, post-injury pain roles, and therapeutic prospects of microglia and astrocytes in neuropathic pain after SCI and elucidates the specific signaling pathways involved. We also speculated that the extracellular matrix (ECM) can affect the glial cells as well. Furthermore, we also mentioned potential targeted therapies, challenges, and progress in clinical trials, as well as new biomarkers and therapeutic targets. Finally, other relevant cell interactions in neuropathic pain and the role of glial cells in other neuropathic pain conditions have been discussed. This review serves as a comprehensive resource for further investigations into the microglia-astrocyte interaction and the detailed mechanisms of neuropathic pain after SCI, with the aim of improving therapeutic efficacy.
Collapse
Affiliation(s)
- Yinuo Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xintong Cai
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Bowen Shi
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yajie Mo
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Jianmin Zhang
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Wenting Luo
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Bodong Yu
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xi Li
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
83
|
Zhuang H, Lei W, Wu Q, Zhao S, Zhao Y, Zhang S, Zhao N, Sun J, Liu Y. Overexpressed CD73 attenuates GSDMD-mediated astrocyte pyroptosis induced by cerebral ischemia-reperfusion injury through the A2B/NF-κB pathway. Exp Neurol 2025; 386:115152. [PMID: 39832662 DOI: 10.1016/j.expneurol.2025.115152] [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] [Revised: 01/03/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Ischemic stroke, resulting from the blockage or narrowing of cerebral vessels, causes brain tissue damage due to ischemia and hypoxia. Although reperfusion therapy is essential to restore blood flow, it may also result in reperfusion injury, causing secondary damage through mechanisms like oxidative stress, inflammation, and excitotoxicity. These effects significantly impact astrocytes, neurons, and endothelial cells, aggravating brain injury and disrupting the blood-brain barrier. CD73, an ectoenzyme that regulates adenosine production through ATP hydrolysis, plays a critical role in purinergic signaling and neuroprotection. During ischemic stroke, CD73 expression is dynamically regulated in response to ischemia and inflammation. It catalyzes the conversion of AMP to adenosine, which activates adenosine receptors to exert neuroprotective effects. Targeting the CD73-adenosine pathway presents a potential therapeutic strategy for mitigating ischemic stroke damage. Pyroptosis, a highly inflammatory form of programmed cell death mediated by inflammasomes like NLRP3 and caspases, plays a significant role in cerebral ischemia-reperfusion injury. Astrocytes, the most abundant CNS cells, contribute to both neuroprotection and injury, with pyroptosis exacerbating inflammation and brain damage. Regulating astrocyte pyroptosis is a promising therapeutic target. Our study investigates CD73's role in regulating astrocyte pyroptosis during ischemia-reperfusion injury. Using CD73 knockout mice and overexpression models, along with in vitro oxygen-glucose deprivation/reperfusion experiments, we found that CD73 overexpression reduces GSDMD-mediated astrocyte pyroptosis via the A2B/NF-κB pathway. These findings offer a novel approach to reducing neuroinflammation, protecting astrocytes, and improving outcomes in ischemic stroke.
Collapse
Affiliation(s)
- Hao Zhuang
- Department of Neurosurgery, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214000, China; Wuxi Medical Center of Nanjing Medical University, Wuxi 214000, China
| | - Wen Lei
- Department of Neurosurgery, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214000, China; Wuxi Medical Center of Nanjing Medical University, Wuxi 214000, China
| | - Qiang Wu
- Department of Neurosurgery, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214000, China; Wuxi Medical Center of Nanjing Medical University, Wuxi 214000, China
| | - Songyun Zhao
- Department of Neurosurgery, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214000, China; Wuxi Medical Center of Nanjing Medical University, Wuxi 214000, China
| | - Yunxuan Zhao
- Department of Endocrinology, Nanjing Hospital of Traditional Chinese Medicine, Nanjing 210001, China
| | - Shizhe Zhang
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Bengbu Medical University, Hefei, Anhui 230001, China
| | - Ning Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Jun Sun
- Department of Neurosurgery, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214000, China; Wuxi Medical Center of Nanjing Medical University, Wuxi 214000, China.
| | - Yuankun Liu
- Department of Neurosurgery, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214000, China; Wuxi Medical Center of Nanjing Medical University, Wuxi 214000, China.
| |
Collapse
|
84
|
Pérez-Núñez R, González MF, Avalos AM, Leyton L. Impacts of PI3K/protein kinase B pathway activation in reactive astrocytes: from detrimental effects to protective functions. Neural Regen Res 2025; 20:1031-1041. [PMID: 38845231 PMCID: PMC11438337 DOI: 10.4103/nrr.nrr-d-23-01756] [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: 10/24/2023] [Revised: 04/07/2024] [Accepted: 05/06/2024] [Indexed: 07/12/2024] Open
Abstract
Astrocytes are the most abundant type of glial cell in the central nervous system. Upon injury and inflammation, astrocytes become reactive and undergo morphological and functional changes. Depending on their phenotypic classification as A1 or A2, reactive astrocytes contribute to both neurotoxic and neuroprotective responses, respectively. However, this binary classification does not fully capture the diversity of astrocyte responses observed across different diseases and injuries. Transcriptomic analysis has revealed that reactive astrocytes have a complex landscape of gene expression profiles, which emphasizes the heterogeneous nature of their reactivity. Astrocytes actively participate in regulating central nervous system inflammation by interacting with microglia and other cell types, releasing cytokines, and influencing the immune response. The phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway is a central player in astrocyte reactivity and impacts various aspects of astrocyte behavior, as evidenced by in silico , in vitro , and in vivo results. In astrocytes, inflammatory cues trigger a cascade of molecular events, where nuclear factor-κB serves as a central mediator of the pro-inflammatory responses. Here, we review the heterogeneity of reactive astrocytes and the molecular mechanisms underlying their activation. We highlight the involvement of various signaling pathways that regulate astrocyte reactivity, including the PI3K/AKT/mammalian target of rapamycin (mTOR), α v β 3 integrin/PI3K/AKT/connexin 43, and Notch/PI3K/AKT pathways. While targeting the inactivation of the PI3K/AKT cellular signaling pathway to control reactive astrocytes and prevent central nervous system damage, evidence suggests that activating this pathway could also yield beneficial outcomes. This dual function of the PI3K/AKT pathway underscores its complexity in astrocyte reactivity and brain function modulation. The review emphasizes the importance of employing astrocyte-exclusive models to understand their functions accurately and these models are essential for clarifying astrocyte behavior. The findings should then be validated using in vivo models to ensure real-life relevance. The review also highlights the significance of PI3K/AKT pathway modulation in preventing central nervous system damage, although further studies are required to fully comprehend its role due to varying factors such as different cell types, astrocyte responses to inflammation, and disease contexts. Specific strategies are clearly necessary to address these variables effectively.
Collapse
Affiliation(s)
- Ramón Pérez-Núñez
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - María Fernanda González
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Ana María Avalos
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
85
|
Lu Y, Zhou R, Zhu R, Wu X, Liu J, Ma Y, Zhang X, Zhang Y, Yang L, Li Y, Zhang Y, Yan Y, Zhang Q. Baicalin ameliorates neuroinflammation by targeting TLR4/MD2 complex on microglia via PI3K/AKT/NF-κB signaling pathway. Neuropharmacology 2025; 267:110296. [PMID: 39798687 DOI: 10.1016/j.neuropharm.2025.110296] [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: 11/26/2024] [Revised: 12/30/2024] [Accepted: 01/04/2025] [Indexed: 01/15/2025]
Abstract
This study aims to elucidate the target and mechanism of baicalin, a clinically utilized drug, in the treatment of neuroinflammatory diseases. Neuroinflammation, characterized by the activation of glial cells and the release of various pro-inflammatory cytokines, plays a critical role in the pathogenesis of various diseases, including spinal cord injury (SCI). The remission of such diseases is significantly dependent on the improvement of inflammatory microenvironment. Toll-like receptor 4/myeloid differentiation protein 2 (TLR4/MD2) complex plays an important role in pathogen recognition and innate immune activation. baicalin, a natural flavonoid, is renowned for its potent anti-inflammatory property. In this study, we discovered that baicalin significantly reduced the activation of glial cells and the levels of pro-inflammatory cytokines at the lesion site of SCI mice, thereby mitigating demyelination and neuronal damage. By directly occupying the active pocket of TLR4/MD2 complex on microglia, baicalin inhibited PI3K/AKT/NF-κB pathway, thereby exerting its anti-inflammatory effect. These findings were corroborated in mice induced by lipopolysaccharide, a TLR4 agonist. Furthermore, baicalin indirectly altered phenotype of astrocytes by reducing secretion of TNF-α, IL-1α, and C1q levels from microglia. Our work demonstrated that baicalin effectively alleviated neuroinflammation by directly targeting microglia and indirectly modulating astrocytes phenotype. As a natural flavonoid, baicalin holds significant potential as a therapeutic candidate for diseases characterized by neuroinflammation.
Collapse
Affiliation(s)
- Yufang Lu
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Ruiying Zhou
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Ruyi Zhu
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Xue Wu
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Xianyang, Shaanxi, 712046, China
| | - Jin Liu
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Yue Ma
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Xin Zhang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Yaling Zhang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Luting Yang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Yanhua Li
- Datong Key Laboratory of Smart Medicine and Health Care for Elderly Chronic Diseases, Medical School, Shanxi Datong University, Datong, Shanxi, 037009, China
| | - Yuan Zhang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Yaping Yan
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China.
| | - Qian Zhang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China.
| |
Collapse
|
86
|
Chen L, Yu Z, Zhu S, Song S, He G, Chi ZL, Wu W. Astrocyte-Derived Extracellular Vesicles Alleviate Optic Nerve Injury Through Remodeling of Retinal Microenvironmental Homeostasis. Invest Ophthalmol Vis Sci 2025; 66:16. [PMID: 40192635 PMCID: PMC11980952 DOI: 10.1167/iovs.66.4.16] [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: 11/08/2024] [Accepted: 03/14/2025] [Indexed: 04/11/2025] Open
Abstract
Purpose Traumatic optic neuropathy (TON) leads to the loss of retinal ganglion cells (RGCs) and results in permanent visual impairment. Protecting and regenerating RGCs is crucial for the treatment of TON. Studies have demonstrated that astrocyte-derived extracellular vesicles (ADEVs) exhibit neuroprotective effects in models of central nervous system (CNS) injury. This study aimed to investigate whether ADEVs have a similar neuroprotective effect on RGCs in an optic nerve crush (ONC) rat model. Methods ADEVs were collected from primary rat astrocytes, and an ONC model was established to evaluate the effects of ADEVs on retinal structure and visual function using optical coherence tomography (OCT), hematoxylin and eosin (H&E) staining, and flash visual evoked potential (f-VEP) analysis. Immunofluorescence was used to examine RGCs and investigate reactive gliotic changes. Additionally, miRNA sequencing of ADEVs and retinal mRNA sequencing were performed to identify the potential mechanisms involved. Results ADEVs protected RGCs from progressive loss and improved visual function. ADEVs also significantly increased the expression of glial fibrillary acidic protein (GFAP) and modulated microglial activation. The miRNAs associated with ADEVs were targeted by neuroprotective signals, such as MAPK, PI3K-AKT, and TNF-α, and through the targeting network generated via retinal mRNA sequencing, we found that potential functional genes, such as THBS1, PAK3, and Gstm1, likely participate in microenvironmental regulation. Conclusions We discovered that ADEVs play a neuroprotective role in optic nerve injury. Our findings provide a new cell-free therapeutic strategy for optic neuropathy.
Collapse
Affiliation(s)
- Lili Chen
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Zhonghao Yu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Senmiao Zhu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Shihan Song
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Guanwen He
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Zai-Long Chi
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Wencan Wu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, China
| |
Collapse
|
87
|
Latchney SE, Raheja AC, Ruiz Lopez BR, Womble PD, Blandin KJ, Lugo JN. Glial changes in the dentate gyrus of neuronal-specific PTEN knockout mice correlate with changes in cell proliferation. J Neuroimmunol 2025; 404:578604. [PMID: 40188528 DOI: 10.1016/j.jneuroim.2025.578604] [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: 02/06/2025] [Revised: 03/25/2025] [Accepted: 03/29/2025] [Indexed: 04/08/2025]
Abstract
Dysregulated hippocampal neurogenesis is a feature of temporal lobe epilepsy (TLE), marked by increased neuronal proliferation. The tumor suppressor gene phosphatase and tensin homolog (PTEN) regulates neuronal proliferation, and its deletion is implicated in TLE. We have previously shown that deletion of neuronal subset-specific (NS)-PTEN in mice increases the number of proliferating cells throughout the dentate gyrus, including subregions that are typically devoid of neurons but rich in glial cells, most notably the Hilus and Molecular Layer. In this study, we hypothesized that NS-PTEN knockout mice would exhibit increased numbers of microglia and astrocytes in these same dentate gyrus subregions. We performed immunohistochemistry for Iba1 (microglia) and GFAP (reactive astrocytes) on wild-type and NS-PTEN knockout mice at 4 and 10 weeks of age. Our data reveal that NS-PTEN knockout mice exhibit increased Iba1+ cell density at both ages, with some male-specific effects. Subregional analysis of the dentate gyrus showed that at 4 weeks, NS-PTEN knockout mice had greater Iba1+ cell density in the Granule Cell Layer (GCL) and Hilus, and at 10 weeks, increases were observed in the GCL, Hilus, and Molecular Layer. Additionally, we observed an increased number of microglia with an amoeboid morphology and fewer with thin, ramified processes. Contrast to Iba1+ microglia, GFAP+ reactive astrocytes were localized to the neurogenic GCL. Importantly, increases in both glial types strongly correlated with heightened cell proliferation (Ki67+ cells), as reported in our previous study, underscoring the role of glial cells in the spatial dysregulation of neurogenesis in NS-PTEN knockout mice.
Collapse
Affiliation(s)
- Sarah E Latchney
- Department of Biology, St. Mary's College of Maryland, St. Mary's City, MD, USA.
| | - Anjali C Raheja
- Department of Biology, St. Mary's College of Maryland, St. Mary's City, MD, USA
| | - Brayan R Ruiz Lopez
- Department of Biology, St. Mary's College of Maryland, St. Mary's City, MD, USA
| | - Paige D Womble
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, USA
| | | | - Joaquin N Lugo
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, USA
| |
Collapse
|
88
|
Tao X, Chen H, Zhu Z, Ren T, Zhen H, Sun X, Song Y, Xu X, Song Z, Liu J. Astrocyte-conditional knockout of MOB2 inhibits the phenotypic conversion of reactive astrocytes from A1 to A2 following spinal cord injury in mice. Int J Biol Macromol 2025; 300:140289. [PMID: 39863205 DOI: 10.1016/j.ijbiomac.2025.140289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
After spinal cord injury (SCI), reactive astrocytes in the injured area are triggered after spinal cord injury (SCI) and to polarize into A1 astrocytes with a proinflammatory phenotype or A2 astrocytes with an anti-inflammatory phenotype. Monopolar spindle binder 2 (MOB2) induces astrocyte stellation, maintains cell homeostasis, and promotes neurite outgrowth; however, its role in the phenotypic transformation of reactive astrocytes remains unclear. Here, we confirmed for the first time that MOB2 is associated with A1/A2 phenotypic switching in reactive astrocytes following SCI in mice. MOB2 modulated A1/A2 transformation in a primary astrocyte reactive cell model. Therefore, we constructed MOB2 conditional knockout mice (MOB2GFAP-CKO) and discovered that conditional knockout of MOB2 inhibited the conversion of reactive astrocytes from A1 to A2 and hindered spinal cord function recovery. Mechanistically, MOB2 increased the activation of PI3K-AKT signaling to promote A1/A2 transformation in vitro, whereas sc79 (an AKT activator) reversed the subtype transformation of reactive astrocytes and improved functional recovery in MOB2GFAP-CKO mice after SCI. Taken together, study provides the first insights into how MOB2 acts as a novel regulator to promote the conversion this of the reactive astrocyte phenotype from A1 to A2, showing great potential for the treatment of SCI.
Collapse
Affiliation(s)
- Xin Tao
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213000, Jiangsu, People's Republic of China; Department of Orthopedics, The People's Hospital of Liyang, Liyang 213300, Jiangsu, People's Republic of China
| | - Haining Chen
- Department of Orthopedics, The First Affiliated Hospital of Jiangsu University, Zhenjiang 212000, Jiangsu, People's Republic of China
| | - Zhenghuan Zhu
- Department of Orthopedics, Changzhou Maternal and Child Health Care Hospital, Changzhou 213000, Jiangsu, People's Republic of China
| | - Tianran Ren
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213000, Jiangsu, People's Republic of China
| | - Hongming Zhen
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213000, Jiangsu, People's Republic of China
| | - Xiaoliang Sun
- Department of Orthopedics, Changzhou Maternal and Child Health Care Hospital, Changzhou 213000, Jiangsu, People's Republic of China
| | - Yu Song
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213000, Jiangsu, People's Republic of China
| | - Xu Xu
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213000, Jiangsu, People's Republic of China
| | - Zhiwen Song
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213000, Jiangsu, People's Republic of China.
| | - Jinbo Liu
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213000, Jiangsu, People's Republic of China.
| |
Collapse
|
89
|
Chen Y, Han L, Zhu DS, Guan YT. Fibrinogen and Neuroinflammation in the Neurovascular Unit in Stroke. J Inflamm Res 2025; 18:4567-4584. [PMID: 40191094 PMCID: PMC11971976 DOI: 10.2147/jir.s496433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 02/13/2025] [Indexed: 04/09/2025] Open
Abstract
Stroke remains a leading cause of death and disability worldwide. Recent evidence suggests that stroke pathophysiology extends beyond vascular dysfunction to include complex interactions within the neurovascular unit (NVU), particularly involving fibrinogen. This blood-derived protein accumulates in the brain following blood-brain barrier (BBB) disruption and plays crucial roles in neuroinflammation and tissue repair. Through its unique structural domains, fibrinogen interacts with multiple cellular components, including astrocytes, microglia, and neural stem cells, thereby modulating inflammatory responses and neural repair mechanisms. This review examines fibrinogen's structure and its diverse functions in stroke pathophysiology, focusing on its interactions with vascular cells, glial cells, and peripheral immune cells. We also discuss emerging therapeutic strategies targeting fibrinogen-mediated pathways and the challenge of translating experimental results into effective clinical treatments.
Collapse
Affiliation(s)
- Yi Chen
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
| | - Lu Han
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
| | - De-Sheng Zhu
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
- Department of Neurology, Baoshan Branch, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200444, People’s Republic of China
| | - Yang-Tai Guan
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
- Department of Neurology, Punan Hospital, Shanghai, 200125, People’s Republic of China
| |
Collapse
|
90
|
Hawkes CH, Giovannoni G, Lechner-Scott J, Levy M, Yeh A. IS MULTIPLE SCLEROSIS A MICROGLIOPATHY? Mult Scler Relat Disord 2025; 96:106403. [PMID: 40174435 DOI: 10.1016/j.msard.2025.106403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Affiliation(s)
- Christopher H Hawkes
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | - Gavin Giovannoni
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | - Michael Levy
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ann Yeh
- Department of Paediatrics (Neurology), Hospital for Sick Children, University of Toronto, Ontario, Canada
| |
Collapse
|
91
|
Zheng T, Marschall S, Weinberg J, Fu X, Tarr A, Shukitt-Hale B, Booth SL. Low Vitamin K Intake Impairs Cognition, Neurogenesis, and Elevates Neuroinflammation in C57BL/6 Mice. J Nutr 2025; 155:1119-1126. [PMID: 39884511 DOI: 10.1016/j.tjnut.2025.01.023] [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: 10/30/2024] [Revised: 01/14/2025] [Accepted: 01/23/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND In addition to its important roles in blood coagulation and bone formation, vitamin K (VK) contributes to brain function. Low dietary VK intake, which is common among older adults, is associated with age-related cognitive impairment. OBJECTIVES To elucidate the biological mechanisms underlying VK's effects on cognition, we investigated the effects of low VK (LVK) intake on cognition in C57BL/6 mice. METHODS Male and female 9-mo-old C57BL/6 mice (n = 60) were fed an LVK diet or a control diet for 6 mo. Behavioral tests were performed on a subset of mice (n = 26) at 15 mo, and brain tissues were collected for follow-up analyses. RESULTS Menaquinone-4, the predominant VK form in the brain, was significantly lower in LVK mice compared to controls (15.6 ± 13.3 compared with 189 ± 186 pmol/g, respectively, P < 0.01). LVK mice showed reduced recognition memory in the novel object test by spending a lower percentage of time exploring the novel object compared to controls (47.45% ± 4.17 compared with 58.08% ± 3.03, P = 0.04). They also spent a significantly longer time learning the task of locating the platform in the Morris water maze test. Within the hippocampal dentate gyrus, LVK mice had a significantly lower number of proliferating cells and fewer newly generated immature neurons compared to control mice. Additionally, more activated microglia cells were identified in the LVK mice. CONCLUSIONS Our data indicate that LVK intake reduced menaquinone-4 concentrations in brain tissues and impaired learning- and memory-related cognitive function. This impairment may be related to the observed reduced hippocampal neurogenesis and elevated neural inflammation.
Collapse
Affiliation(s)
- Tong Zheng
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States.
| | - Shannon Marschall
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Jasper Weinberg
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Xueyan Fu
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Andrew Tarr
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Barbara Shukitt-Hale
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Sarah L Booth
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| |
Collapse
|
92
|
Xue X, Zhang Z, Yang Y, Hu L, Zhao M, He Q. Acute exposure of perchlorate on zebrafish larvae: Neurotoxicity during development. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 294:118111. [PMID: 40158377 DOI: 10.1016/j.ecoenv.2025.118111] [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: 10/18/2024] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
OBJECTIVE This study aimed to assess the acute developmental toxicity of perchlorate exposure and identify its manifestation of neurotoxicity in zebrafish (Danio rerio) model. METHODS Zebrafish larvae were exposed to sodium perchlorate at early developmental stages and the phenotypes of various developmental abnormalities were observed and recorded. Neurodevelopmental toxicity has been studied in particular in terms of central nervous system apoptosis, peripheral motor abnormalities, and abnormal autonomic motor behavior. Samples were processed for pathological, transcriptomic, and PCR analyses. RESULTS Acute exposure of zebrafish larvae to perchlorate resulted in developmental toxicity in a concentration-dependent manner. Heart rate abnormality (33 %), slow blood flow (37 %), abnormal size of liver (23 %), delayed yolk absorption (70 %), and abnormal body pigmentation (100 %) were observed in the 3.83 mg/mL group. Heart edema (10 %), heart rate abnormality (60 %), slow blood flow (67 %), abnormal size of liver (77 %), delayed yolk absorption (100 %), abnormalities in intestinal morphology (30 %), abnormal body pigmentation (100 %), and shorter body length (47 %) were observed in the 4.03 mg/mL group. Developmental neurotoxicity was characterized by brain apoptosis, damage to the central nervous system (P < 0.01) and peripheral motor neurons (P < 0.0001), and reduced autonomous motor distance (P < 0.01). Transcriptomic analysis revealed that c3a.1, c5, fga, fgb, and fgg were upregulated in the complement and coagulation cascades, and the mRNA levels of c3a.1, dusp1, slc2a6, purba, and klf9 were found upregulated in PCR. CONCLUSION Acute perchlorate exposure on zebrafish larvae caused various developmental toxicity in a concentration-dependent manner, notably neurotoxicity. We believed the immunological and inflammatory responses were involved.
Collapse
Affiliation(s)
- Xiaoran Xue
- Department of Pediatrics, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Zaiqiu Zhang
- Department of Pediatrics, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Yang Yang
- Department of Pediatrics, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Lin Hu
- Department of Pediatrics, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Mingyi Zhao
- Department of Pediatrics, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Qingnan He
- Department of Pediatrics, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
93
|
Kim Y, Vaidya B, McInnes J, Zoghbi HY. Alpha-Synuclein Phosphomimetic Y39E and S129D Knock-In Mice Show Cytosolic Alpha-Synuclein Localization without Developing Neurodegeneration or Motor Deficits. eNeuro 2025; 12:ENEURO.0357-24.2025. [PMID: 40164505 PMCID: PMC11998964 DOI: 10.1523/eneuro.0357-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 04/02/2025] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by motor and nonmotor symptoms. Its pathological hallmarks include the accumulation of misfolded alpha-synuclein (α-Syn) in Lewy bodies and Lewy neurites. Phosphorylation of α-Syn is a prominent feature of these inclusions, but its role in disease pathogenesis remains unclear. To identify the role of α-Syn phosphorylation in synucleinopathy, we generated two Snca knock-in (KI) mouse models carrying phosphomimetic mutations at SncaY39 or SncaS129 (SncaY39E or SncaS129D ) which manipulated epitopes phosphorylated in the PD brain. Both SncaY39E and SncaS129D KI mice displayed increased α-Syn phosphorylation, enhanced oligomer formation, and a shift of α-Syn localization from membrane-bound to cytoplasm. However, neurodegeneration in the substantia nigra was not observed up to 24 months of age. These findings demonstrate that mimicking the phosphorylation of Y39 or S129 can induce endogenous α-Syn phosphorylation. Still, a single phosphomimetic mutation alone is insufficient to induce PD-like behavior and pathology in the mouse's lifespan. Overall, our study provides a mouse model for investigating the role of phosphorylation at Y39 and S129 α-Syn epitopes in vivo.
Collapse
Affiliation(s)
- YoungDoo Kim
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030
| | - Bhupesh Vaidya
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030
| | - Joseph McInnes
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030
| | - Huda Y Zoghbi
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas 77030
- Department of Neuroscience, BCM, Houston, Texas 77030
- Department of Pediatrics, BCM, Houston, Texas 77030
- Department of Neurology, BCM, Houston, Texas 77030
- Howard Hughes Medical Institute, Houston, Texas 77030
| |
Collapse
|
94
|
Wu Z, Xu L, Xie Y, Sambangi A, Swaminathan S, Pei Z, Ji W, Li Z, Guo Y, Li Z, Chen G. Brain-Wide Neuroregenerative Gene Therapy Improves Cognition in a Mouse Model of Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410080. [PMID: 39951299 PMCID: PMC11984881 DOI: 10.1002/advs.202410080] [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: 08/22/2024] [Revised: 01/09/2025] [Indexed: 04/12/2025]
Abstract
Alzheimer's disease (AD) is a progressive and irreversible brain disorder with extensive neuronal loss in the neocortex and hippocampus. Current therapeutic interventions focus on the early stage of AD but lack effective treatment for the late stage of AD, largely due to the inability to replenish the lost neurons and repair the broken neural circuits. In this study, by using engineered adeno-associated virus vectors that efficiently cross the blood-brain-barrier in the mouse brain, a brain-wide neuroregenerative gene therapy is developed to directly convert endogenous astrocytes into functional neurons in a mouse model of AD. It is found that ≈500 000 new neurons are regenerated and widely distributed in the cerebral cortex and hippocampus. Importantly, it is demonstrated that the converted neurons can integrate into pre-existing neural networks and improve various cognitive performances in AD mice. Chemogenetic inhibition of the converted neurons abolishes memory enhancement in AD mice, suggesting a pivotal role for the newly converted neurons in cognitive restoration. Together, brain-wide neuroregenerative gene therapy may provide a viable strategy for the treatment of AD and other brain disorders associated with massive neuronal loss.
Collapse
Affiliation(s)
- Zheng Wu
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐Human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
- Department of BiologyHuck Institutes of Life SciencesPennsylvania State UniversityUniversity ParkPA16802USA
| | - Liang Xu
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐Human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
| | - Yu Xie
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐Human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
| | - Abhijeet Sambangi
- Department of BiologyHuck Institutes of Life SciencesPennsylvania State UniversityUniversity ParkPA16802USA
| | - Shreya Swaminathan
- Department of BiologyHuck Institutes of Life SciencesPennsylvania State UniversityUniversity ParkPA16802USA
| | - Zifei Pei
- Department of BiologyHuck Institutes of Life SciencesPennsylvania State UniversityUniversity ParkPA16802USA
| | - Wenyu Ji
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐Human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
| | - Zeru Li
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐Human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
| | - Yaowei Guo
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐Human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
| | - Zhifei Li
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐Human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
| | - Gong Chen
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐Human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
- Department of BiologyHuck Institutes of Life SciencesPennsylvania State UniversityUniversity ParkPA16802USA
| |
Collapse
|
95
|
Diep YN, Park HJ, Zhu X, Nam JH, Shim JK, Chang JH, Nguyen DD, Kim I, Jo DG, Lee LP, Yun M, Cho H. A Neuroimmune-Oncology Microphysiological Analysis Platform (NEO-MAP) for Evaluating Astrocytic Scar Formation and Microgliosis in Glioblastoma Microenvironment. Adv Healthc Mater 2025; 14:e2404821. [PMID: 40072333 DOI: 10.1002/adhm.202404821] [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: 02/28/2025] [Indexed: 04/26/2025]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of brain tumor, characterized by its heterogeneity in cellular components, including reactive astrocytes and microglia. Since neuroimmune responses like astrogliosis and microgliosis gain recognition as vital factors in brain tumor progression, there is a growing need for clinically relevant models that assess the interactions between astrocytes, microglia, and GBM. Here, a NEuroimmune-Oncology Microphysiological Analysis Platform (NEO-MAP) is presented as a "new map" to observe astrocytic scar formation and microgliosis in response to GBM. NEO-MAP based on pathophysiological principles is designed to replicate the GBM-glia interactions, multi-phenotypic microglia activities, scar-forming astrocytes with chondroitin sulfate proteoglycans (CSPGs) in the extracellular matrix, and the biophysical characteristics of the astrocytic scar barrier. The NEO-MAP reveals that inhibiting mTORC2 in GBM promotes the proinflammatory transformation of astrocytes and enhanced astrocytic scar formation. Astrocytes that form scars prompted microglia to change from the M2 to M1 phenotype, enhancing chemotherapy sensitivity. Tissues from GBM patients show a significant correlation between reduced mTORC2 activity and increased astrogliosis, alongside a decrease in M2-polarized microgliosis, aligning with the NEO-MAP findings. Overall, the NEO-MAP is foreseen as a clinically significant tool for exploring tumor-glia interactions, opening avenues for drug development aimed at the tumor microenvironment.
Collapse
Affiliation(s)
- Yen N Diep
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hee Jung Park
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Xiaohui Zhu
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jin Ho Nam
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jin-Kyoung Shim
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Brain Tumor Translational Research Laboratory, Severance Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Dang Du Nguyen
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Inki Kim
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Dong-Gyu Jo
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Biomedical Institute for Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, Republic of Korea
| | - Luke P Lee
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Harvard Medical School, Division of Engineering in Medicine and Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, 94720, USA
| | - Mijin Yun
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hansang Cho
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
96
|
Dhar A, Moinuddin FM, Zamanian CA, Sharar AD, Dominari A, Graepel S, Windebank AJ, Bydon M. SOX Genes in Spinal Cord Injury: Redefining Neural Stem Cell Regeneration Strategies. Mol Neurobiol 2025:10.1007/s12035-025-04882-w. [PMID: 40156684 DOI: 10.1007/s12035-025-04882-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
The study design is literature review. The sex-determining region Y gene (SRY)-related high mobility group box (HMG)-box (SOX) gene family has primarily been associated with neural development and sex determination and is a key component of human embryonic development. Recent studies on zebrafish models have demonstrated that the unique ability of the latter for central nervous tissue (CNS) repair following injury is largely mediated by SOX genes. Given that efforts aimed at the structural regeneration and functional restoration of neural tissue still represent a major therapeutic challenge in patients suffering CNS injury, these findings have initiated a discussion regarding the development of novel therapeutic strategies for SCI focusing on neural tissue regeneration. Spinal cord injury (SCI), in particular, represents a field that could greatly benefit from studies related to the function of the SOX genes. Neuro-informatics Laboratory, Mayo Clinic, Rochester, MN. A literature review was conducted, with a focus on SOX gene that has been described in the experimental studies of SCI. In this review, the existing evidence linking the SOX gene family to the pathophysiology of SCI is summarized, and future research steps regarding the potential implications of the SOX genes in neurological recovery following SCI are discussed, especially focusing on highlighting potential therapeutic targets. The potential implications of the latter could play a crucial role in future efforts to advance the treatment approaches to SCI.
Collapse
Affiliation(s)
- Ashis Dhar
- Mayo Clinic Neuro-Informatics Laboratory, Department of Neurosurgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | - F M Moinuddin
- Mayo Clinic Neuro-Informatics Laboratory, Department of Neurosurgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | - Cameron A Zamanian
- Mayo Clinic Neuro-Informatics Laboratory, Department of Neurosurgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | - Ahnaf Dil Sharar
- Mayo Clinic Neuro-Informatics Laboratory, Department of Neurosurgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | - Asimina Dominari
- Mayo Clinic Neuro-Informatics Laboratory, Department of Neurosurgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | - Stephen Graepel
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Mohamad Bydon
- Mayo Clinic Neuro-Informatics Laboratory, Department of Neurosurgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
97
|
Zayed M, Kim YC, Jeong BH. Biological characteristics and transcriptomic profile of adipose-derived mesenchymal stem cells isolated from prion-infected murine model. Stem Cell Res Ther 2025; 16:154. [PMID: 40156048 PMCID: PMC11951670 DOI: 10.1186/s13287-025-04273-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: 03/22/2024] [Accepted: 03/11/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Prion diseases are characterized by accumulation of misfolded host prion proteins (PrPSc) that produce aggregates in brain tissue. Mesenchymal stem cells (MSCs) have been identified as potential therapeutic candidates for prion diseases. However, it has been demonstrated that MSCs maintained and expressed PrPSc levels following inoculation, raising concerns regarding their safe and effective use in medical applications. Prion infectivity has been reported in fat tissues, thus the response of adipose-derived MSCs (AdMSCs) to prion infection needs to be fully studied. METHODS For this study, we analyzed the properties of AdMSCs isolated from mice infected with the ME7 scrapie strain and compared them with negative controls. We investigated morphology, viability, immunophenotyping, markers of inflammation, migration activity, and neurotrophic factors. RNA sequencing (RNA-Seq) was performed to identify transcriptome profile changes. RESULTS AdMSCs derived from ME7-infected mice displayed immunophenotypes similar to cells from negative controls, but they were larger with lower viability (p < 0.05). ME7 infection caused higher expression of inflammatory mediators CCL5, TNF-α, C3, and IL6 (p < 0.05 and p < 0.01) and low expression of the stem cell marker, CXCR4 (p < 0.05) which was confirmed by immunofluorescence staining. The results showed decreased migration activity and wound closure ability of AdMSCs isolated from ME7-infected mice as confirmed by Transwell migration and scratch wound assays (p < 0.05 and p < 0.001), respectively. The RNA-Seq results detected 367 differentially expressed genes between AdMSCs from ME7-infected mice and those from the negative controls, and negative regulation of locomotion, extracellular matrix (ECM) organization, collagen-containing ECM, and extracellular structure organization genes were common in AdMSCs from ME7-infected mice. Transcriptomic analysis revealed that pathways enriched in AdMSCs from ME7-infected mice included those involved in the PI3K-Akt signaling pathway, cell adhesion, protein digestion and absorption, and cytokine-cytokine receptor interactions. Interestingly, genes related to the regulation of iron storage, such as Hp and hepcidin, were upregulated in AdMSCs isolated from ME7-infected mice. CONCLUSIONS Based on these data, therapeutic strategies for AdMSCs in prion disease should be further investigated.
Collapse
Affiliation(s)
- Mohammed Zayed
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, 54531, Republic of Korea
- Department of Bioactive Material Sciences and Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju, 54896, Republic of Korea
- Department of Surgery, College of Veterinary Medicine, South Valley University, Qena, 83523, Egypt
| | - Yong-Chan Kim
- Department of Biological Sciences, Andong National University, Andong, 36729, Republic of Korea
- School of Life Sciences and Biotechnology, Gyeongkuk National University, Andong 36729, Republic of Korea
| | - Byung-Hoon Jeong
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, 54531, Republic of Korea.
- Department of Bioactive Material Sciences and Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju, 54896, Republic of Korea.
| |
Collapse
|
98
|
Batsuuri K, Toychiev AH, Viswanathan S, Wohl SG, Srinivas M. Targeting Connexin 43 in Retinal Astrocytes Promotes Neuronal Survival in Glaucomatous Injury. Glia 2025. [PMID: 40156150 DOI: 10.1002/glia.70013] [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: 08/23/2024] [Revised: 03/04/2025] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
Astrocytes in the retina and optic nerve head play an important role in the pathogenesis of glaucoma. Astrocytes extensively express connexin 43 (Cx43), a protein that forms gap junction (GJ) channels and transmembrane unopposed hemichannels. While it is well documented that Cx43 expression is augmented in retinal injuries, the role of astrocytic Cx43 channels in glaucomatous injury is not fully understood. Here, we used a mouse model of ocular hypertension caused by intracameral microbead injections and a more severe model, optic nerve crush (ONC) injury, and assessed changes in Cx43 expression and GJ channel function. The effect of astrocyte-specific deletion of Cx43 (Cx43KO) on retinal ganglion cell (RGC) loss and visual function was also assessed. We show that the Cx43 expression is increased in retinal astrocytes at early time points and remained elevated even after sustained elevation of intraocular pressure (IOP) (~8 weeks), which paralleled an increase in astrocytic GJ coupling. Deletion of astrocytic Cx43 markedly improved the survival of RGCs by ~93% and preserved visual function as assessed by ERG and reduced numbers of activated microglial/macrophages in the glaucomatous retina. Cx43 expression was also substantially increased after ONC injury, and the absence of Cx43 in this model increased RGC survival by ~48%. These results reveal a deleterious role for Cx43 in glaucoma progression. Intravitreal injections of Gap19, a peptide that reportedly inhibits Cx43 hemichannels but not GJ channels, markedly increased RGC survival and visual function. Further studies are required to assess whether targeting Cx43 hemichannels might be useful for glaucoma treatment.
Collapse
Affiliation(s)
- Khulan Batsuuri
- Department of Biological and Vision Sciences, SUNY College of Optometry, New York, New York, USA
| | - Abduqodir H Toychiev
- Department of Biological and Vision Sciences, SUNY College of Optometry, New York, New York, USA
| | | | - Stefanie G Wohl
- Department of Biological and Vision Sciences, SUNY College of Optometry, New York, New York, USA
| | - Miduturu Srinivas
- Department of Biological and Vision Sciences, SUNY College of Optometry, New York, New York, USA
| |
Collapse
|
99
|
Martinez MX, Mahler SV. Potential roles for microglia in drug addiction: Adolescent neurodevelopment and beyond. J Neuroimmunol 2025; 404:578600. [PMID: 40199197 DOI: 10.1016/j.jneuroim.2025.578600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/14/2025] [Accepted: 03/27/2025] [Indexed: 04/10/2025]
Abstract
Adolescence is a sensitive period for development of addiction-relevant brain circuits, and it is also when people typically start experimenting with drugs. Unfortunately, such substance use may cause lasting impacts on the brain, and might increase vulnerability to later-life addictions. Microglia are the brain's immune cells, but their roles in shaping neural connectivity and synaptic plasticity, especially in developmental sensitive periods like adolescence, may also contribute to addiction-related phenomena. Here, we overview how drugs of abuse impact microglia, and propose that they may play poorly-understood, but important roles in addiction vulnerability and progression.
Collapse
Affiliation(s)
- Maricela X Martinez
- Department of Neurobiology and Behavior, University of California, 2221 McGaugh Hall, Irvine, CA 92697, USA.
| | - Stephen V Mahler
- Department of Neurobiology and Behavior, University of California, 2221 McGaugh Hall, Irvine, CA 92697, USA
| |
Collapse
|
100
|
Hu J, Xie S, Chen T, Liao Y, Qian Z, Zhang L. Glial vascular Unit as a bridge between Blood-Brain Barrier and glymphatic System: Roles in sepsis-associated encephalopathy. Neuroscience 2025; 570:68-71. [PMID: 39984027 DOI: 10.1016/j.neuroscience.2025.02.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/02/2025] [Accepted: 02/17/2025] [Indexed: 02/23/2025]
Abstract
This article underscores the Glial Vascular Unit (GVU) 's possible role in bridging the Blood-Brain Barrier (BBB) and Glymphatic System in Sepsis-associated encephalopathy (SAE). Future studies should prioritize understanding the mechanistic underpinnings of GVU dysfunction in sepsis and explore interventions aimed at modulating BBB permeability, astrocytic function, and glymphatic clearance. Understanding these complex mechanisms is crucial for developing therapeutic strategies aimed at mitigating the neurological impact of sepsis and improving outcomes for patients with SAE.
Collapse
Affiliation(s)
- Jiyun Hu
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Shucai Xie
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Tao Chen
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Ya Liao
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Zhaoxin Qian
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| | - Lina Zhang
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| |
Collapse
|