1
|
Claeys W, Geerts A, Van Hoecke L, Van Steenkiste C, Vandenbroucke RE. Role of astrocytes and microglia in hepatic encephalopathy associated with advanced chronic liver disease: lessons from animal studies. Neural Regen Res 2025; 20:3461-3475. [PMID: 39688562 PMCID: PMC11974659 DOI: 10.4103/nrr.nrr-d-24-00600] [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: 05/28/2024] [Revised: 08/05/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Hepatic encephalopathy, defined as neuropsychiatric dysfunction secondary to liver disease, is a frequent decompensating event in cirrhosis. Its clinical impact is highlighted by a notable increase in patient mortality rates and a concomitant reduction in overall quality of life. Systemically, liver disease, liver function failure, portosystemic shunting, and associated multi-organ dysfunction result in the increase of disease-causing neurotoxins in the circulation, which impairs cerebral homeostasis. Key circulating neurotoxins are ammonia and inflammatory mediators. In the brain, pathophysiology is less well understood, but is thought to be driven by glial cell dysfunction. Astrocytes are the only brain resident cells that have ammonia-metabolizing machinery and are therefore putatively most susceptible to ammonia elevation. Based on a large body of mostly in vitro evidence, ammonia-induced cellular and molecular disturbances include astrocyte swelling and oxidative stress. Microglia, the brain resident macrophages, have been linked to the translation of systemic inflammation to the brain microenvironment. Recent evidence from animal studies has provided novel insights into old and new downstream effects of astrocyte and microglial dysfunction such as toxin clearance disruption and myeloid cell attraction to the central nervous system parenchyma. Furthermore, state of the art research increasingly implicates neuronal dysfunction and possibly even irreversible neuronal cell death. Cell-type specific investigation in animal models highlights the need for critical revision of the contribution of astrocytes and microglia to well-established and novel cellular and molecular alterations in hepatic encephalopathy. In this review, we therefore give a current and comprehensive overview of causes, features, and consequences of astrocyte and microglial dysfunction in hepatic encephalopathy, including areas of interest for future investigation.
Collapse
Affiliation(s)
- Wouter Claeys
- Department of Internal Medicine and Paediatrics, Hepatology Research Unit, Ghent University, Ghent, Belgium
- Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium
- Barriers in Inflammation, VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Anja Geerts
- Department of Internal Medicine and Paediatrics, Hepatology Research Unit, Ghent University, Ghent, Belgium
- Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium
- Department of Gastroenterology and Hepatology, Ghent University Hospital, Ghent, Belgium
| | - Lien Van Hoecke
- Barriers in Inflammation, VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Christophe Van Steenkiste
- Department of Gastroenterology and Hepatology, Antwerp University, Antwerp, Belgium
- Department of Gastroenterology and Hepatology, Maria Middelares Hospital, Ghent, Belgium
| | - Roosmarijn E. Vandenbroucke
- Barriers in Inflammation, VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
2
|
Jovanovic MZ, Stanojevic J, Stevanovic I, Ninkovic M, Ilic TV, Nedeljkovic N, Dragic M. Prolonged intermittent theta burst stimulation restores the balance between A2AR- and A1R-mediated adenosine signaling in the 6-hydroxidopamine model of Parkinson's disease. Neural Regen Res 2025; 20:2053-2067. [PMID: 39254566 PMCID: PMC11691459 DOI: 10.4103/nrr.nrr-d-23-01542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 04/30/2024] [Accepted: 06/17/2024] [Indexed: 09/11/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202507000-00027/figure1/v/2024-09-09T124005Z/r/image-tiff An imbalance in adenosine-mediated signaling, particularly the increased A2AR-mediated signaling, plays a role in the pathogenesis of Parkinson's disease. Existing therapeutic approaches fail to alter disease progression, demonstrating the need for novel approaches in PD. Repetitive transcranial magnetic stimulation is a non-invasive approach that has been shown to improve motor and non-motor symptoms of Parkinson's disease. However, the underlying mechanisms of the beneficial effects of repetitive transcranial magnetic stimulation remain unknown. The purpose of this study is to investigate the extent to which the beneficial effects of prolonged intermittent theta burst stimulation in the 6-hydroxydopamine model of experimental parkinsonism are based on modulation of adenosine-mediated signaling. Animals with unilateral 6-hydroxydopamine lesions underwent intermittent theta burst stimulation for 3 weeks and were tested for motor skills using the Rotarod test. Immunoblot, quantitative reverse transcription polymerase chain reaction, immunohistochemistry, and biochemical analysis of components of adenosine-mediated signaling were performed on the synaptosomal fraction of the lesioned caudate putamen. Prolonged intermittent theta burst stimulation improved motor symptoms in 6-hydroxydopamine-lesioned animals. A 6-hydroxydopamine lesion resulted in progressive loss of dopaminergic neurons in the caudate putamen. Treatment with intermittent theta burst stimulation began 7 days after the lesion, coinciding with the onset of motor symptoms. After treatment with prolonged intermittent theta burst stimulation, complete motor recovery was observed. This improvement was accompanied by downregulation of the eN/CD73-A2AR pathway and a return to physiological levels of A1R-adenosine deaminase 1 after 3 weeks of intermittent theta burst stimulation. Our results demonstrated that 6-hydroxydopamine-induced degeneration reduced the expression of A1R and elevated the expression of A2AR. Intermittent theta burst stimulation reversed these effects by restoring the abundances of A1R and A2AR to control levels. The shift in ARs expression likely restored the balance between dopamine-adenosine signaling, ultimately leading to the recovery of motor control.
Collapse
Affiliation(s)
- Milica Zeljkovic Jovanovic
- Laboratory for Neurobiology, Department of General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Jelena Stanojevic
- Medical Faculty of Military Medical Academy, University of Defence, Belgrade, Serbia
| | - Ivana Stevanovic
- Medical Faculty of Military Medical Academy, University of Defence, Belgrade, Serbia
| | - Milica Ninkovic
- Medical Faculty of Military Medical Academy, University of Defence, Belgrade, Serbia
| | - Tihomir V. Ilic
- Medical Faculty of Military Medical Academy, University of Defence, Belgrade, Serbia
| | - Nadezda Nedeljkovic
- Laboratory for Neurobiology, Department of General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Milorad Dragic
- Laboratory for Neurobiology, Department of General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
3
|
González-Velasco O, Simon M, Yilmaz R, Parlato R, Weishaupt J, Imbusch C, Brors B. Identifying similar populations across independent single cell studies without data integration. NAR Genom Bioinform 2025; 7:lqaf042. [PMID: 40276039 PMCID: PMC12019640 DOI: 10.1093/nargab/lqaf042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/13/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025] Open
Abstract
Supervised and unsupervised methods have emerged to address the complexity of single cell data analysis in the context of large pools of independent studies. Here, we present ClusterFoldSimilarity (CFS), a novel statistical method design to quantify the similarity between cell groups across any number of independent datasets, without the need for data correction or integration. By bypassing these processes, CFS avoids the introduction of artifacts and loss of information, offering a simple, efficient, and scalable solution. This method match groups of cells that exhibit conserved phenotypes across datasets, including different tissues and species, and in a multimodal scenario, including single-cell RNA-Seq, ATAC-Seq, single-cell proteomics, or, more broadly, data exhibiting differential abundance effects among groups of cells. Additionally, CFS performs feature selection, obtaining cross-dataset markers of the similar phenotypes observed, providing an inherent interpretability of relationships between cell populations. To showcase the effectiveness of our methodology, we generated single-nuclei RNA-Seq data from the motor cortex and spinal cord of adult mice. By using CFS, we identified three distinct sub-populations of astrocytes conserved on both tissues. CFS includes various visualization methods for the interpretation of the similarity scores and similar cell populations.
Collapse
Affiliation(s)
- Oscar González-Velasco
- Division Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Division of Neurodegenerative Disorders, Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, 68167 Mannheim, Germany
| | - Malte Simon
- Division Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Leibniz Institute for Immunotherapy, 93053 Regensburg, Germany
| | - Rüstem Yilmaz
- Division of Neurodegenerative Disorders, Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, 68167 Mannheim, Germany
| | - Rosanna Parlato
- Division of Neurodegenerative Disorders, Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, 68167 Mannheim, Germany
| | - Jochen Weishaupt
- Division of Neurodegenerative Disorders, Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, 68167 Mannheim, Germany
| | - Charles D Imbusch
- Division Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Institute of Immunology, University Medical Center Mainz, 55131 Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, 55131 Mainz, Germany
| | - Benedikt Brors
- Division Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Medical Faculty Heidelberg and Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
4
|
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] [MESH Headings] [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
|
5
|
Seplovich G, Bouchi Y, de Rivero Vaccari JP, Pareja JCM, Reisner A, Blackwell L, Mechref Y, Wang KK, Tyndall JA, Tharakan B, Kobeissy F. Inflammasome links traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease. Neural Regen Res 2025; 20:1644-1664. [PMID: 39104096 PMCID: PMC11688549 DOI: 10.4103/nrr.nrr-d-24-00107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/20/2024] [Accepted: 06/03/2024] [Indexed: 08/07/2024] Open
Abstract
Traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease are three distinct neurological disorders that share common pathophysiological mechanisms involving neuroinflammation. One sequela of neuroinflammation includes the pathologic hyperphosphorylation of tau protein, an endogenous microtubule-associated protein that protects the integrity of neuronal cytoskeletons. Tau hyperphosphorylation results in protein misfolding and subsequent accumulation of tau tangles forming neurotoxic aggregates. These misfolded proteins are characteristic of traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease and can lead to downstream neuroinflammatory processes, including assembly and activation of the inflammasome complex. Inflammasomes refer to a family of multimeric protein units that, upon activation, release a cascade of signaling molecules resulting in caspase-induced cell death and inflammation mediated by the release of interleukin-1β cytokine. One specific inflammasome, the NOD-like receptor protein 3, has been proposed to be a key regulator of tau phosphorylation where it has been shown that prolonged NOD-like receptor protein 3 activation acts as a causal factor in pathological tau accumulation and spreading. This review begins by describing the epidemiology and pathophysiology of traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease. Next, we highlight neuroinflammation as an overriding theme and discuss the role of the NOD-like receptor protein 3 inflammasome in the formation of tau deposits and how such tauopathic entities spread throughout the brain. We then propose a novel framework linking traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease as inflammasome-dependent pathologies that exist along a temporal continuum. Finally, we discuss potential therapeutic targets that may intercept this pathway and ultimately minimize long-term neurological decline.
Collapse
Affiliation(s)
| | - Yazan Bouchi
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jennifer C. Munoz Pareja
- Division of Pediatric Critical Care, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andrew Reisner
- Department of Pediatrics, Emory University, Atlanta, GA, USA
- Department of Neurosurgery, Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Laura Blackwell
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA
| | - Kevin K. Wang
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | | | - Binu Tharakan
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| |
Collapse
|
6
|
Liss A, Siddiqi MT, Marsland P, Varodayan FP. Neuroimmune regulation of the prefrontal cortex tetrapartite synapse. Neuropharmacology 2025; 269:110335. [PMID: 39904409 DOI: 10.1016/j.neuropharm.2025.110335] [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: 11/14/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
The prefrontal cortex (PFC) is an essential driver of cognitive, affective, and motivational behavior. There is clear evidence that the neuroimmune system directly influences PFC synapses, in addition to its role as the first line of defense against toxins and pathogens. In this review, we first describe the core structures that form the tetrapartite PFC synapse, focusing on the signaling microdomain created by astrocytic cradling of the synapse as well as the emerging role of the extracellular matrix in synaptic organization and plasticity. Neuroimmune signals (e.g. pro-inflammatory interleukin 1β) can impact the function of each core structure within the tetrapartite synapse, as well as promote intra-synaptic crosstalk, and we will provide an overview of recent advances in this field. Finally, evidence from post mortem human brain tissue and preclinical studies indicate that inflammation may be a key contributor to PFC dysfunction. Therefore, we conclude with a mechanistic discussion of neuroimmune-mediated maladaptive plasticity in neuropsychiatric disorders, with a focus on alcohol use disorder (AUD). Growing recognition of the neuroimmune system's role as a critical regulator of the PFC tetrapartite synapse provides strong support for targeting the neuroimmune system to develop new pharmacotherapeutics.
Collapse
Affiliation(s)
- Andrea Liss
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - Mahum T Siddiqi
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - Paige Marsland
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - Florence P Varodayan
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA.
| |
Collapse
|
7
|
Shvachiy L, Amaro-Leal Â, Machado F, Rocha I, Geraldes V, Outeiro TF. Lead as an environmental toxicant in models of synucleinopathies. CHEMOSPHERE 2025; 380:144477. [PMID: 40347673 DOI: 10.1016/j.chemosphere.2025.144477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 04/08/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Lead, a toxic heavy metal, is prevalent in various industrial applications, contributing to environmental contamination and significant health concerns. Lead affects various body systems, especially the brain, causing long-lasting cognitive and behavioral changes. While most studies have focused on continuous lead exposure, intermittent exposure, such as that caused by migration or relocations, has received less attention. Importantly, lead exposure intensifies the severity of Parkinson's disease (PD) and dementia with Lewy bodies, diseases involving the accumulation of alpha-synuclein (aSyn) in the brain and in the gut. Although the precise mechanisms underlying these observations remain unclear, oxidative stress and mitochondrial dysfunction likely play a role. Here, we investigated how two different profiles of lead exposure - continuous and intermittent - affect models of synucleinopathies. We found that lead exposure enhances the formation of aSyn inclusions, resulting in an increase in both their number and size in cell models. In addition, we found that animals injected with aSyn pre-formed fibrils display serine 129-phosphorylated aSyn inclusions and a reduction in astrocytes in the substantia nigra. These animals also display neuronal damage and alterations in locomotor activity, exploration behavior, anxiety, memory impairments and hypertension. Our results suggest a mechanistic link between environmental lead exposure and the onset and progression of diseases associated with aSyn pathology. Understanding the molecular and cellular interactions between lead and aSyn is crucial for shaping public health policies and may provide novel insight into strategies for mitigating the impact of environmental toxins on neurodegenerative processes involved in Parkinson's disease and related synucleinopathies.
Collapse
Affiliation(s)
- Liana Shvachiy
- University Medical Center Göttingen, Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, Göttingen, 37073, Germany; Cardiovascular Centre of the University of Lisbon, Lisbon, 1649-028, Portugal; Institute of Physiology, Faculty of Medicine of the University of Lisbon, Portugal
| | - Ângela Amaro-Leal
- Cardiovascular Centre of the University of Lisbon, Lisbon, 1649-028, Portugal; Institute of Physiology, Faculty of Medicine of the University of Lisbon, Portugal; Egas Moniz School of Health and Science, 2829-511, Caparica, Portugal
| | - Filipa Machado
- Cardiovascular Centre of the University of Lisbon, Lisbon, 1649-028, Portugal
| | - Isabel Rocha
- Cardiovascular Centre of the University of Lisbon, Lisbon, 1649-028, Portugal; Institute of Physiology, Faculty of Medicine of the University of Lisbon, Portugal
| | - Vera Geraldes
- Cardiovascular Centre of the University of Lisbon, Lisbon, 1649-028, Portugal; Institute of Physiology, Faculty of Medicine of the University of Lisbon, Portugal.
| | - Tiago F Outeiro
- University Medical Center Göttingen, Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, Göttingen, 37073, Germany; Max Planck Institute for Muldisciplinary Science, 37075, Göttingen, Germany; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK; Scientific Employee with an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany.
| |
Collapse
|
8
|
Chen J, Xu S, Wang L, Liu X, Liu G, Tan Q, Li W, Zhang S, Du Y. Refining the interactions between microglia and astrocytes in Alzheimer's disease pathology. Neuroscience 2025; 573:183-197. [PMID: 40120713 DOI: 10.1016/j.neuroscience.2025.03.033] [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: 12/06/2024] [Revised: 03/03/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Microglia and astrocytes are central to the pathogenesis and progression of Alzheimer's Disease (AD), working both independently and collaboratively to regulate key pathological processes such as β-amyloid protein (Aβ) deposition, tau aggregation, neuroinflammation, and synapse loss. These glial cells interact through complex molecular pathways, including IL-3/IL-3Ra and C3/C3aR, which influence disease progression and cognitive decline. Emerging research suggests that modulating these pathways could offer therapeutic benefits. For instance, recombinant IL-3 administration in mice reduced Aβ plaques and improved cognitive functions, while C3aR inhibition alleviated Aβ and tau pathologies, restored synaptic function, and corrected immune dysregulation. However, the effects of these interactions are context-dependent. Acute C3/C3aR activation enhances microglial Aβ clearance, whereas chronic activation impairs it, highlighting the dual roles of glial signaling in AD. Furthermore, C3/C3aR signaling not only impacts Aβ clearance but also modulates tau pathology and synaptic integrity. Given AD's multifactorial nature, understanding the specific pathological environment is crucial when investigating glial cell contributions. The interplay between microglia and astrocytes can be both neuroprotective and neurotoxic, depending on the disease stage and brain region. This complexity underscores the need for targeted therapies that modulate glial cell activity in a context-specific manner. By elucidating the molecular mechanisms underlying microglia-astrocyte interactions, this research advances our understanding of AD and paves the way for novel therapeutic strategies aimed at mitigating neurodegeneration and cognitive decline in AD and related disorders.
Collapse
Affiliation(s)
- Jiangmin Chen
- College of Acupuncture-Moxibustion and Orthopaedics, Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China
| | - Shuyu Xu
- College of Acupuncture-Moxibustion and Orthopaedics, Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China
| | - Li Wang
- College of Acupuncture-Moxibustion and Orthopaedics, Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China
| | - Xinyuan Liu
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Guangya Liu
- College of Acupuncture-Moxibustion and Orthopaedics, Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China
| | - Qian Tan
- College of Acupuncture-Moxibustion and Orthopaedics, Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China
| | - Weixian Li
- College of Acupuncture-Moxibustion and Orthopaedics, Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China
| | - Shuai Zhang
- College of Acupuncture-Moxibustion and Orthopaedics, Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China
| | - Yanjun Du
- College of Acupuncture-Moxibustion and Orthopaedics, Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China; Hubei Shizhen Laboratory, China; Hubei International Science and Technology Cooperation Base of Preventive Treatment by Acupuncture and Moxibustion, China; Hubei Provincial Hospital of Traditional Chinese Medicine, China.
| |
Collapse
|
9
|
Nowaczewska-Kuchta A, Ksiazek-Winiarek D, Glabinski A. Interaction Between Neutrophils and Elements of the Blood-Brain Barrier in the Context of Multiple Sclerosis and Ischemic Stroke. Int J Mol Sci 2025; 26:4437. [PMID: 40362673 PMCID: PMC12072651 DOI: 10.3390/ijms26094437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 05/03/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025] Open
Abstract
The blood-brain barrier (BBB) is a semi-permeable membrane in physiological conditions, but in pathologies like multiple sclerosis (MS) and ischemic stroke (IS), its permeability increases. In this review, we focus on neutrophils and their interaction with cellular components of the BBB: endothelial cells (EC), pericytes (PC), and astrocytes (AC). Nowadays, neutrophils receive more attention, mostly due to advanced research techniques that show the complexity of their population. Additionally, neutrophils have the ability to secrete extracellular vesicles (EVs), reactive oxygen species (ROS) and cytokines, which both destroy and restore the BBB. Astrocytes, PCs, and ECs also have dual roles in the pathogenesis of MS and IS. The interaction between neutrophils and cellular components of the BBB provides us with a wider insight into the pathogenesis of common diseases in the central nervous system. Further, we comprehensively review knowledge about the influence of neutrophils on the BBB in the context of MS and IS. Moreover, we describe new therapeutic strategies for patients with MS and IS like cell-based therapies and therapies that use the neutrophil function.
Collapse
Affiliation(s)
| | | | - Andrzej Glabinski
- Department of Neurology and Stroke, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland; (A.N.-K.); (D.K.-W.)
| |
Collapse
|
10
|
Tong X, Liu X, Jiang YX, Su JR, Luan JQ, Guo C. Astrocyte lactoferrin deficiency affects the construction and function of spinal neurons by regulating cholesterol metabolism. Exp Cell Res 2025; 449:114595. [PMID: 40334811 DOI: 10.1016/j.yexcr.2025.114595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/27/2025] [Accepted: 05/05/2025] [Indexed: 05/09/2025]
Abstract
Astrocytes play pivotal roles in central nervous system (CNS) homeostasis, with emerging evidence implicating astrocyte-derived lactoferrin (Lf) in neurodevelopmental and neurodegenerative processes. This study investigates Lf's functional significance in spinal cord integrity using astrocyte-specific Lf knockout (cKO) mice. Behavioral analyses of 1-month-old male cKO mice revealed impaired motor coordination (increased balance beam scores and prolonged pole-climbing latency) and delayed nociceptive responses (increased thermal withdrawal latency). Morphological assessments demonstrated neuron-specific pathology: motor neurons exhibited atrophy and reduced Nissl substance staining, spinal ganglion cells showed quantitative depletion with vacuolar degeneration, and protein expression analyses confirmed declines in neuronal markers (NeuN), synaptic components (SNAP25, PSD95), axonal and myelin related proteins (NF-L, MBP), and neurotransmitter transporters (AChE). Notably, glial cell populations remained unaffected. Mechanistic investigations identified reduced spinal cholesterol content accompanied by downregulation of cholesterol biosynthesis and transport regulators (Srebp2, HMGCR, ApoE, ABCA1) and activation of AMP-activated protein kinase (AMPK). These findings establish astrocytic Lf as a critical modulator of cholesterol metabolism essential for maintaining neuronal structural and functional integrity in the spinal cord. The discovered Lf-cholesterol regulatory axis provides novel insights into the pathogenesis of spinal cord disorders and highlights potential therapeutic targets for neurodegenerative conditions.
Collapse
Affiliation(s)
- Xin Tong
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Xin Liu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Yu-Xuan Jiang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Jia-Rui Su
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Jun-Qi Luan
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Chuang Guo
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China.
| |
Collapse
|
11
|
Chauhan P, Begum MY, Narapureddy BR, Gupta S, Wadhwa K, Singh G, Kumawat R, Sharma N, Ballal S, Jha SK, Abomughaid MM, B D, Ojha S, Jha NK. Unveiling the Involvement of Herpes Simplex Virus-1 in Alzheimer's Disease: Possible Mechanisms and Therapeutic Implications. Mol Neurobiol 2025; 62:5850-5874. [PMID: 39648189 DOI: 10.1007/s12035-024-04535-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/01/2024] [Indexed: 12/10/2024]
Abstract
Viruses pose a significant challenge and threat to human health, as demonstrated by the current COVID-19 pandemic. Neurodegeneration, particularly in the case of Alzheimer's disease (AD), is significantly influenced by viral infections. AD is a neurodegenerative disease that affects people of all ages and poses a significant threat to millions of individuals worldwide. The precise mechanism behind its development is not yet fully understood; however, the emergence and advancement of AD can be hastened by various environmental factors, such as bacterial and viral infections. There has been a longstanding suspicion that the herpes simplex virus-1 (HSV-1) may have a role to play in the development or advancement of AD. Reactivation of HSV-1 could potentially lead to damage to neurons, either by direct means or indirectly by triggering inflammation. This article provides an overview of the connection between HSV-1 infections and immune cells (astrocytes, microglia, and oligodendrocytes) in the progression of AD. It summarizes recent scientific research on how HSV-1 affects neurons, which could potentially shed light on the clinical features and treatment options for AD. In addition, the paper has explored the impact of HSV-1 on neurons and its role in various aspects of AD, such as Aβ secretion, tau hyperphosphorylation, metabolic dysregulation, oxidative damage, apoptosis, and autophagy. It is believed that the immune response triggered by HSV-1 reactivation plays a role in the development of neurodegeneration in AD. Despite the lack of a cure for AD, researchers have made significant efforts to study the clinical and pathological aspects of the disease, identify biomarkers, and gain insight into its underlying causes. The goal is to achieve early diagnosis and develop treatments that can modify the progression of the disease. The current article discusses the most promising therapy for combating the viral impacts, which provides additional evidence for the frequent reactivations of latent HSV-1 in the AD brain. However, further research is still required to establish the molecular and cellular mechanisms underlying the development of AD through the reactivation of HSV-1. This could potentially lead to new insights in drug development aimed at preventing HSV-1 reactivation and the subsequent development and progression of AD.
Collapse
Affiliation(s)
- Payal Chauhan
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - M Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Bayapa Reddy Narapureddy
- Department of Public Health, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Karan Wadhwa
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - Govind Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India.
| | - Rohit Kumawat
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajsthan, Jaipur, India
| | - Naveen Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges Jhanjeri, Mohali, 140307, Punjab, India
| | - Suhas Ballal
- Departmant of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, Delhi, 110008, India
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Dheepak B
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, United Arab Emirates
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Biosciences & Technology, Galgotias University, Greater Noida, India.
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India.
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, 144411, India.
| |
Collapse
|
12
|
Cen YY, Gao XL, Feng YH, Zhou C, Li CJ, Liu F, Shen JF, Zhang YY. The Double-Edged Effect of Connexins and Pannexins of Glial Cells in Central and Peripheral Nervous System After Nerve Injury. Mol Neurobiol 2025:10.1007/s12035-025-04991-6. [PMID: 40310549 DOI: 10.1007/s12035-025-04991-6] [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: 01/25/2025] [Accepted: 04/21/2025] [Indexed: 05/02/2025]
Abstract
Glial cells play pivotal roles in homeostatic regulation and driving reactive pathologic changes after nerve injury. Connexins (Cxs) and pannexins (Panxs) have emerged as seminal proteins implicated in cell-cell communication, exerting a profound impact on the response processes of glial cell activation, proliferation, protein synthesis and secretion, as well as apoptosis following nerve injury. These influences are mediated through various forms, including protein monomers, hemichannel (HC), and gap junction (GJ), mainly by regulating intercellular or intracellular signaling pathways. Multiple Cx and Panx isoforms have been detected in central nervous system (CNS) or peripheral nervous system (PNS). Each isoform exhibits distinct cellular and subcellular localization, and the differential regulation and functional roles of various protein isoforms are observed post-injury. The quantitative and functional alterations of the same protein isoform in different studies remain inconsistent, attributable to factors such as the predominant mode of protein polymerization, the specific injury model, and the injury site. Similarly, the same protein isoforms have different roles in regulating the response processes after nerve injury, thus exerting a double-edged sword effect. This review describes the regulatory mechanisms and bidirectional effects of Cxs and Panxs. Additionally, it surveys the current status of research and application of drugs as therapeutic targets for neuropathic injuries. We summarize comprehensive and up-to-date information on these proteins in the glial cell response to nerve injury, providing new perspectives for future mechanistic exploration and development of targeted therapeutic approaches.
Collapse
Affiliation(s)
- Yue-Yan Cen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
| | - Xin-Lin Gao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
| | - Yu-Heng Feng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
| | - Cheng Zhou
- Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu, China
| | - Chun-Jie Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fei Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
| | - Jie-Fei Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China.
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China.
| | - Yan-Yan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China.
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China.
| |
Collapse
|
13
|
Wang Y, Wu Y, Wang Y, Xiong R, Ling C, Cao Y, Wang Y, Yang Y, Qu Z, Xu N, Liu S, Li W, Lv Z, Hu Z, Fan C. CVA16 infection causes neurological injury by engaging TLR2/MYD88/TNF-α/CXCL1 signalling pathway in hSCARB2 knock-in mice. Antiviral Res 2025; 237:106133. [PMID: 40054503 DOI: 10.1016/j.antiviral.2025.106133] [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: 02/15/2025] [Accepted: 02/27/2025] [Indexed: 03/15/2025]
Abstract
Coxsackievirus A16 (CVA16), a major pathogen responsible for hand-foot-and-mouth disease (HFMD) in children, has frequently replaced Enterovirus A71 as the predominant causative agent in China and other Asia-Pacific regions. The lack effective drugs and vaccines against this virus exacerbates the concerns on its outbreaks. Clinical reports and laboratory studies indicate that CVA16 infection may lead to neurological injury, but the precise mechanisms remain elusive. In this study, we meticulously established a CVA16 murine disease model using 3-week-old hSCARB2 knock-in mice through intracranial inoculation. Within 4-7 days post-infection, the infected mice exhibited severe neurological symptoms featured as limb paralysis, hind limb weakness and ataxia. Furthermore, high viral loads were detected in the brain, spleen, skeletal muscle tissues, indicating a systemic infection. A robust cytokine response was observed, characterized with the elevation of TNF-α, IL-12 (p40), IL-10 and MIP-1β. Histological and immunofluorescence staining revealed extensive inflammation, marked by the concentrated infiltration of astrocytes cells, as well as severe neurological injury, which included hypertrophic and extended pseudopodia microglia, increased astrocytes with long and stretched protuberances, markedly decreased neuronal cell bodies and nerve fibers in brain. No visible pathological changes were observed in spinal cord tissues. RNA sequencing and immunofluorescence staining of brain tissue verification assays indicated that the neurological injury may engage in TLR2/MYD88/TNF-α/CXCL1 signal pathway. Over all, this work addressed the gap in the availability of CVA16 disease rodent model for vaccine development and provided novel insights into the mechanisms underlying neurological injury caused by enteroviruses and other neurotropic viruses.
Collapse
Affiliation(s)
- Yu Wang
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Yong Wu
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Yuya Wang
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Rui Xiong
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Chen Ling
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Yuan Cao
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Yining Wang
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Yanwei Yang
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Zhe Qu
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Nan Xu
- Institute for Biological Products Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Susu Liu
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Weijia Li
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Zhe Lv
- Sinovac Biotech Ltd., Beijing, China
| | - Zhongyu Hu
- Institute for Biological Products Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China.
| | - Changfa Fan
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, China.
| |
Collapse
|
14
|
Ouard N, Tali A, Souhoudji TD, Jebbouj R, El-Bouchikhi I, Rose CF, Ahboucha S. Different cortical and subcortical astroglial responsiveness in rats with acute liver failure. J Neuropathol Exp Neurol 2025; 84:412-422. [PMID: 40173416 DOI: 10.1093/jnen/nlaf020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025] Open
Abstract
Hepatic encephalopathy (HE) is a neuropsychiatric complication of liver failure. Previous studies described astroglia alterations in HE, but regional changes have not been well investigated. This study addresses regional astroglial response by exploring glial fibrillary acidic protein (GFAP) immunoreactivity in cortical structures including somatosensory (S1Tr and S1BF), piriform (Pir), and perirhinal (PRh) cortices, and subcortical regions including corpus callosum (CC), ventromedial thalamus (VMT), mammillothalamic tract (MTT), and dorsomedial hypothalamic nucleus (DHN) in rats with acute liver failure (ALF) sacrificed at coma stage. Our data showed decreased numbers of astrocytes in S1Tr, Pir, and CC in ALF rats. GFAP-immunoreactive cells were increased within other regions including PRh, VMT, MTT, and DHN. Cell morphometric analysis showed significant increase in GFAP-immunoreactive astrocyte processes and cell bodies in cortical and subcortical regions but not in CC and DHN. However, astrocyte perimeters were increased, particularly in S1Tr and VMT. Our study demonstrates regional specificity including (1) regions with astrocyte activation associated with an increase of GFAP-immunostaining and astrocyte cell counts, together with (2) unaltered GFAP components, and (3) regions characterized by presumably inactive astrocyte with a reduced GFAP-immunostaining. These findings may reflect either different regional alterations in HE, or stages of an alteration progressing differently in different regions.
Collapse
Affiliation(s)
- Nahla Ouard
- Multidisciplinary Laboratory of Research and Innovation (MLRI), Technological Applications, Environmental Resources and Health Research Team (ATRES), Polydisciplinary Faculty, Sultan Moulay Slimane University, Khouribga, Morocco
| | - Assmaâ Tali
- Multidisciplinary Laboratory of Research and Innovation (MLRI), Technological Applications, Environmental Resources and Health Research Team (ATRES), Polydisciplinary Faculty, Sultan Moulay Slimane University, Khouribga, Morocco
| | - Themoi Demsou Souhoudji
- Multidisciplinary Laboratory of Research and Innovation (MLRI), Technological Applications, Environmental Resources and Health Research Team (ATRES), Polydisciplinary Faculty, Sultan Moulay Slimane University, Khouribga, Morocco
| | - Rajâa Jebbouj
- Multidisciplinary Laboratory of Research and Innovation (MLRI), Technological Applications, Environmental Resources and Health Research Team (ATRES), Polydisciplinary Faculty, Sultan Moulay Slimane University, Khouribga, Morocco
| | - Ihssane El-Bouchikhi
- Multidisciplinary Laboratory of Research and Innovation (MLRI), Technological Applications, Environmental Resources and Health Research Team (ATRES), Polydisciplinary Faculty, Sultan Moulay Slimane University, Khouribga, Morocco
| | - Christopher F Rose
- Hepato-Neuro Lab, CRCHUM, Montréal, Canada
- Médicine Département, Université de Montréal, Montréal, Canada
| | - Samir Ahboucha
- Multidisciplinary Laboratory of Research and Innovation (MLRI), Technological Applications, Environmental Resources and Health Research Team (ATRES), Polydisciplinary Faculty, Sultan Moulay Slimane University, Khouribga, Morocco
| |
Collapse
|
15
|
Wood LB, Singer AC. Neurons as Immunomodulators: From Rapid Neural Activity to Prolonged Regulation of Cytokines and Microglia. Annu Rev Biomed Eng 2025; 27:55-72. [PMID: 39805040 DOI: 10.1146/annurev-bioeng-110122-120158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Regulation of the brain's neuroimmune system is central to development, normal function, and disease. Neuronal communication to microglia, the primary immune cells of the brain, is well known to involve purinergic signaling mediated via ATP secretion and the cytokine fractalkine. Recent evidence shows that neurons release multiple cytokines beyond fractalkine, yet these are less studied and poorly understood. In contrast to ATP, cytokines are a class of signaling molecule that are much larger, with longer signaling and farther diffusion. We posit that neuron-expressed cytokines are an essential mechanism of neuron-microglia communication that arises as part of both normal learning and memory and in response to tissue pathology. Thus, neurons are underappreciated immunomodulatory cells that express diverse immunomodulatory signals. While neuronally sourced cytokines have been understudied, new technical advances make this a timely topic. The goal of this review is to define what is known about the cytokines expressed from neurons, how they are regulated, and the effects of these cytokines on microglia. We delineate key knowledge gaps and needs for new tools to define and analyze neuronal roles in immunomodulation. Given that cytokines are central regulators of microglial function, a broad new body of work is required to illuminate functional links between these neuronally expressed cytokines and sustained and transient microglial function.
Collapse
Affiliation(s)
- Levi B Wood
- Wallace H. Coulter Department of Biomedical Engineering, George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA; ,
| | - Annabelle C Singer
- Wallace H. Coulter Department of Biomedical Engineering, George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA; ,
| |
Collapse
|
16
|
Peyton L, Haroon H, Umpierre A, Essa H, Bruce R, Wu LJ, Choi DS. In vivo calcium extrusion from accumbal astrocytes reduces anxiety-like behaviors but increases compulsive-like responses and compulsive ethanol drinking in mice. Neuropharmacology 2025; 268:110320. [PMID: 39842625 PMCID: PMC11830519 DOI: 10.1016/j.neuropharm.2025.110320] [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: 12/22/2023] [Revised: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
The ventral striatum is crucially involved in reward processing. The present study investigates the behavioral effects of astrocyte-specific calcium extrusion virus "CalEx" on perseverative responses in the operant five-choice serial reaction time task and ethanol-conditioned place preference. Mice were injected with CalEx via the GfaABC1D promoter to extrude cytosolic calcium from astrocytes within the ventral striatum. We found that CalEx transfection in the ventral striatum reduced evoked response duration, the maximum amplitude, and the response frequency to 500 μM ATP as measured by ΔF/F fluorescence intensity of the genetically encoded calcium indicator targeting astrocytes GCaMP6f. During the five-choice serial reaction time task, CalEx mice persisted in perseverative responses compared to their counterparts. Additionally, during ethanol-conditioned place preference, CalEx mice showed increased place preference for a low ethanol concentration compared to control group. Furthermore, we found that accumbal astrocytic calcium extrusion increased quinine adulterated ethanol drinking. Our findings suggest that diminishing ventral striatum astrocyte calcium activity contributes to compulsive behaviors, ethanol drinking, and enhanced ethanol drug reward.
Collapse
Affiliation(s)
- Lee Peyton
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | - Humza Haroon
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | | | - Hesham Essa
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | - Robert Bruce
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA; Neuroscience Program, Mayo Clinic College of Medicine and Science, MN, 55905, USA; Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA; Neuroscience Program, Mayo Clinic College of Medicine and Science, MN, 55905, USA; Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, 59905, USA.
| |
Collapse
|
17
|
Geier B, Roy B, Reiter LT. Small molecule ion channel agonist/antagonist screen reveals seizure suppression via glial Irk2 activation in a Drosophila model of Dup15q syndrome. Neurobiol Dis 2025; 208:106882. [PMID: 40122181 DOI: 10.1016/j.nbd.2025.106882] [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: 01/22/2025] [Revised: 03/20/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025] Open
Abstract
The neurogenetic disorder duplication 15q syndrome (Dup15q) is characterized by a high incidence of autism spectrum disorder (ASD) and pharmacoresistant epilepsy. Standard-of-care broad-spectrum anti-seizure medications (ASM) often fail to control seizures in Dup15q, emphasizing the need for the identification of new therapeutic compounds. Previously, we generated a model of Dup15q in Drosophila melanogaster by overexpressing Dube3a in glial cells, instead of neurons. This model recapitulates the spontaneous seizures present in Dup15q patients. Here, we screened a set of FDA-approved compounds for their ability to suppress seizures in repo > Dube3a flies. We used 72 compounds from the Enzo SCREEN-WELL Ion Channel Library for primary screening of seizure suppression. Six compounds were identified that significantly reduced seizure duration. Furthermore, the compounds that passed the primary and secondary screenings were associated with K+ channels. Glial-specific knockdown of the inward rectifying potassium (Irk) 2 channel exacerbated the seizure phenotype in these animals indicating a mechanism of action for drugs that bind irk2, like minoxidil, and can suppress seizures through the rebalancing of K+ extracellularly. This pharmacological and molecular investigation further supports the role of extracellular K+ content in Dup15q seizure activation and provides a putative target for therapeutic intervention.
Collapse
Affiliation(s)
- Benjamin Geier
- Department of Physiology, Tulane University, New Orleans, LA, USA; Graduate Program in Neuroscience, Tulane University, New Orleans, LA, USA
| | - Bidisha Roy
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA
| | | |
Collapse
|
18
|
Verkhratsky A, Li B, Niu J, Lin SS, Su Y, Jin WN, Li Y, Jiang S, Yi C, Shi FD, Tang Y. Neuroglial Advances: New Roles for Established Players. J Neurochem 2025; 169:e70080. [PMID: 40371609 DOI: 10.1111/jnc.70080] [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: 03/29/2025] [Accepted: 04/17/2025] [Indexed: 05/16/2025]
Abstract
Neuroglial cells perform numerous physiological functions and contribute to the pathogenesis of all diseases of the nervous system. Neuroglial neuroprotection defines the resilience of the nervous tissue to exo- and endogenous pathological challenges, while neuroglial defence determines the progression and outcome of neurological disorders. IN this paper, we overview previously unknown but recently discovered roles of various types of neuroglial cells in diverse physiological and pathological processes. First, we describe the role of ependymal glia in the regulation of cerebrospinal fluid flow from the spinal cord to peripheral tissues through the spinal nerves. This newly discovered pathway provides a highway for the CNS-body volume transmission. Next, we present the mechanism by which astrocytes control migration and differentiation of oligodendrocyte precursor cells (OPCs). In pre- and early postnatal CNS, OPCs migrate using vasculature (which is yet free from glia limitans perivascularis) as a pathfinder. Newly forming astrocytic perivascular endfeet signal (through semaphorin-plexin cascade) to OPCs that detach from the vessels and start to differentiate into myelinating oligodendrocytes. We continue the astrocyte theme by demonstrating the neuroprotective role of APOE-laden astrocytic extracellular vesicles in neuromyelitis optica. Next, we explore the link between astrocytic morphology and stress-induced depression. We discuss the critical role of astrocytic ezrin, the cytosolic linker defining terminal astrocyte arborisation and resilience to stress: overexpression of ezrin in prefrontal cortical astrocytes makes mice resistant to stress, whereas ezrin knockdown increases animals vulnerability to stress. Subsequently, we highlight the pathophysiological role of oligodendroglial lineage in schizophrenia by describing novel hypertrophied OPCs in the post-mortem patient's tissue and in a mouse model with OPCs overexpressing alternative splice variant DISC1-Δ3. These DISC1-Δ3-OPCs demonstrated overactivated Wnt/β-catenin signalling pathway and were sufficient to trigger pathological behaviours. Finally, we deliberate on the pathological role of astrocytic and microglial connexin 43 hemichannels in Alzheimer's disease and present a new formula of Cx43 hemichannel inhibitor with increased blood-brain barrier penetration and brain retention.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- International Joint Research Centre on Purinergic Signalling of Sichuan Province, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Department of Neurosciences, University of the Basque Country, Leioa, Bizkaia, Spain
- IKERBASQUE Basque Foundation for Science, Bilbao, Spain
- Celica, BIOMEDICAL, Technology Park 24, Ljubljana, Slovenia
| | - Baoman Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
| | - Jianqin Niu
- Department of Histology and Embryology, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - Si-Si Lin
- International Joint Research Centre on Purinergic Signalling of Sichuan Province, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yixun Su
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Wei-Na Jin
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yifan Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
| | - Shihe Jiang
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chenju Yi
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Shenzhen, China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, China
| | - Fu-Dong Shi
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yong Tang
- International Joint Research Centre on Purinergic Signalling of Sichuan Province, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
19
|
Wang S, Yang Y, Lin J, Zhang W, Yang C, Zhang R, Zhou C, Zhang L, Wang X, Liu J, Jin X, Ma Y. Astragalin actives autophagy and inhibits apoptosis of astrocytes in AD mice via down-regulating Fas/Fasl-VDAC1 pathway. Free Radic Biol Med 2025; 232:72-85. [PMID: 40032030 DOI: 10.1016/j.freeradbiomed.2025.02.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/05/2025]
Abstract
Alzheimer's disease (AD) as a common neurodegenerative disease, which characterized by amyloid Aβ deposition and neurofibrillary tangles. Astragalin (AST), a natural flavonoid, has anti-inflammatory, antioxidant, anti-cancer, and other pharmacological effects. Astrocytes can phagocytize and degrade Aβ in their vicinity. In this study, we used the AD mice model established by injecting the mixture of Aβ1-42 and Aβ25-35 into the CA1 region of the hippocampus, and C8D1A cells injured by Aβ1-42 to explore the neuroprotective effects of AST. Our findings showed that AST enhances learning and cognitive ability of AD mice, reduces Aβ deposition and neurofibrillary tangles in the brain, and improves the structural morphology of hippocampal nerve cells. Furthermore, AST promoted autophagy and suppressed apoptosis of astrocytes in the AD model. Additionally, AST inhibited the expression of proteins associated with the Fas/Fasl-VDAC1 signaling pathway, while autophagy inhibitor chloroquine (CQ) or apoptosis agonist phenoxodiol reversed above change. Interestingly, consistent with the action of pathway Fas inhibitor KR-33493, AST could activate autophagy of Aβ1-42 injured C8D1A cells while inhibit their apoptosis. In conclusion, AST activated autophagy and inhibited apoptosis of hippocampal astrocytes in AD mice, ameliorating animal cognitive deficits by down-regulating Fas/Fasl-VDAC1 signaling pathway. Thus, this study provided a new perspective and experimental foundation for developing AD treatment drugs.
Collapse
Affiliation(s)
- Shuhan Wang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yaqi Yang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jiahong Lin
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Weishan Zhang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Cuizhu Yang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Runheng Zhang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Chang Zhou
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Li Zhang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xin Wang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jing Liu
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaobao Jin
- Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuxin Ma
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
20
|
De Simone U, Caloni F, Pignatti P, Gaetano C, Locatelli CA, Coccini T. Human stromal cell-based protocol to generate astrocytes: a straightforward in vitro predictive strategy in neurotoxicology. Toxicol Mech Methods 2025; 35:340-355. [PMID: 39626968 DOI: 10.1080/15376516.2024.2435351] [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: 09/09/2024] [Revised: 11/22/2024] [Accepted: 11/23/2024] [Indexed: 05/04/2025]
Abstract
The inherent adaptability of human mesenchymal stromal cells (hMSCs) to differentiate into neural lineages provides a valuable resource for investigating potential neurotoxicity in humans. By harnessing the ability of hMSCs to transform into astrocytes, we can evaluate the effects of various agents on these vital cells. Our protocol employs hMSCs sourced from umbilical cord tissue, ensuring a readily available supply of high-quality cells. The hMSC-to-neural workflow encompasses six essential steps: hMSC culture, followed by the generation of embryoid bodies (EBs) from these cells on specialized surfaces. Next, EBs and cells are expanded in a growth-promoting medium, directing them toward neural lineages. Subsequent differentiation into immature astrocytes is achieved through the use of specific factors. The process continues with the maturation of EBs/cells into astrocyte-like cells (hALCs) under optimized conditions, culminating in the final development of hALCs in a specialized medium. This methodology yields cells that display astrocyte morphology and express characteristic markers such as GFAP and S100β. The protocol is efficient, requiring roughly 6 weeks to generate hALCs from primary hMSCs without genetic manipulation. The application of hMSCs in evaluating cell damage triggered by neurotoxicants like MeHg and MGO underscores their potential as a valuable component within a more extensive battery of neurotoxicity tests.
Collapse
Affiliation(s)
- Uliana De Simone
- Laboratory of Clinical and Experimental Toxicology, and Pavia Poison Center-National Toxicology Information Center, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Francesca Caloni
- Dipartimento di Scienze e Politiche Ambientali (ESP), Università degli Studi di Milano, Milan, Italy
| | - Patrizia Pignatti
- Allergy and Immunology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Carlo Gaetano
- Laboratory of Epigenetics, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Carlo Alessandro Locatelli
- Laboratory of Clinical and Experimental Toxicology, and Pavia Poison Center-National Toxicology Information Center, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Teresa Coccini
- Laboratory of Clinical and Experimental Toxicology, and Pavia Poison Center-National Toxicology Information Center, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| |
Collapse
|
21
|
Rezagholizadeh N, Datta G, Hasler WA, Nguon EC, Smokey EV, Chen X. TLR7 Mediates HIV-1 Tat-Induced Cellular Senescence in Human Astrocytes. Aging Cell 2025:e70086. [PMID: 40304459 DOI: 10.1111/acel.70086] [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: 10/31/2024] [Revised: 03/11/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Cellular senescence contributes to accelerated aging, neuroinflammation, and the development of HIV-associated neurocognitive disorders (HAND) in the era of combined antiretroviral therapy (cART). One HIV viral factor that could lead to cellular senescence is the persistence of HIV-1 Tat in the brain. As a secreted viral protein, Tat is known to enter endolysosomes of cells through receptor-mediated endocytosis, and we have shown that Tat induces endolysosome damage and dysfunction. Significantly, endolysosome dysfunction has been strongly linked to cellular senescence. However, it is not known whether endolysosome dysfunction represents a driver or consequence of cellular senescence. Because Tat-induced endolysosome damage represents an early step in exogenous Tat-induced cellular senescence, we tested the hypothesis that Tat induces cellular senescence via an endolysosome-dependent mechanism in human astrocytes. We demonstrated that Tat interacts with an endolysosome-resident Toll-like receptor 7 (TLR7) via its arginine-rich basic domain, and such an interaction results in endolysosome damage and the development of a senescence-like phenotype including cell cycle arrest, enhanced SA-β-gal activity, and increased release of senescence-associated secretory phenotype (SASP) factors (IL-6, IL-8, and CCL2). Thus, our finding provided mechanistic insights whereby Tat induces endolysosome damage and cellular senescence in human astrocytes. We provide compelling evidence that endolysosome damage drives the development of cellular senescence. Our findings also highlight the novel role of TLR7 in the development of cellular senescence and suggest that TLR7 represents a novel therapeutic target against senescence and the development of HAND.
Collapse
Affiliation(s)
- Neda Rezagholizadeh
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, USA
| | - Gaurav Datta
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, USA
| | - Wendie A Hasler
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, USA
| | - Erica C Nguon
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, USA
| | - Elise V Smokey
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, USA
| | - Xuesong Chen
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, USA
| |
Collapse
|
22
|
Gaur P, Vaibhav K, Ahluwalia M, Gupta S. Editorial: Neuro-immune interplay: unraveling the complexities of neurological complications and immunology. Front Mol Neurosci 2025; 18:1607675. [PMID: 40370576 PMCID: PMC12075194 DOI: 10.3389/fnmol.2025.1607675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/16/2025] Open
Affiliation(s)
- Pankaj Gaur
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States
| | - Kumar Vaibhav
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Meenakshi Ahluwalia
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Seema Gupta
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
23
|
Wang S, Baumert R, Séjourné G, Sivadasan Bindu D, Dimond K, Sakers K, Vazquez L, Moore JL, Tan CX, Takano T, Rodriguez MP, Brose N, Bradley L, Lessing R, Soderling SH, La Spada AR, Eroglu C. PD-linked LRRK2 G2019S mutation impairs astrocyte morphology and synapse maintenance via ERM hyperphosphorylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.04.09.536178. [PMID: 39253496 PMCID: PMC11383028 DOI: 10.1101/2023.04.09.536178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Astrocytes are highly complex cells that mediate critical roles in synapse formation and maintenance by establishing thousands of direct contacts with synapses through their perisynaptic processes. Here, we found that the most common Parkinsonism gene mutation, LRRK2 G2019S, enhances the phosphorylation of the ERM proteins (Ezrin, Radixin, and Moesin), components of the perisynaptic astrocyte processes in a subset of cortical astrocytes. The ERM hyperphosphorylation was accompanied by decreased astrocyte morphological complexity and reduced excitatory synapse density and function. Dampening ERM phosphorylation levels in LRRK2 G2019S mouse astrocytes restored both their morphology and the excitatory synapse density in the anterior cingulate cortex. To determine how LRRK2 mutation impacts Ezrin interactome, we used an in vivo BioID proteomic approach, and we found that astrocytic Ezrin interacts with Atg7, a master regulator of autophagy. The Ezrin/Atg7 interaction is inhibited by Ezrin phosphorylation, thus diminished in LRRK2 G2019S astrocytes. Importantly, the Atg7 function is required to maintain proper astrocyte morphology. Our data provide a molecular pathway through which the LRRK2 G2019S mutation alters astrocyte morphology and synaptic density in a brain-region-specific manner.
Collapse
|
24
|
Beretta C, Dakhel A, Eltom K, Rosqvist F, Uzoni S, Mothes T, Fletcher JS, Risérus U, Sehlin D, Rostami J, Michno WP, Erlandsson A. Astrocytic lipid droplets contain MHCII and may act as cogs in the antigen presentation machinery. J Neuroinflammation 2025; 22:117. [PMID: 40275347 PMCID: PMC12023685 DOI: 10.1186/s12974-025-03452-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/19/2025] [Indexed: 04/26/2025] Open
Abstract
Lipid droplets (LDs) are crucial for energy homeostasis, but are also involved in a wide spectrum of other cellular processes. Accumulating data identifies LDs as an important player in inflammation. However, the underlying mechanisms and the impact of LDs on neuroinflammation remain unclear. Here, we describe a novel function of LDs in human astrocytes, in the context of Alzheimer's disease (AD). Although, the overall lipid profile was unchanged in astrocytes with AD pathology, our data show a clear effect on LD metabolism and specific fatty acids involved in neuroinflammation. Importantly, we found astrocytes to be in close contact with infiltrating CD4 + T cells in the AD brain. Moreover, PLIN3 + LDs in astrocytes co-localize with major histocompatibility complex II (MHCII), indicating a role of LDs in adaptive immunity. Comprehensive analysis of human induced pluripotent stem cell (hiPSC)-derived astrocytes revealed that MHCII is in fact loaded within PLIN3 + LDs and forwarded to neighboring cells via tunneling nanotubes and secretion. Notably, the MHCII molecules are cleaved into its active form prior to packing, indicating an alternative route of MHCII shuttling through LDs, transporting functional immune complexes between cells. Quantification of PLIN3 + LDs in astrocytic cultures, human brain tissue and cerebral organoids indicates that AD pathology initially stimulates PLIN3 + LD formation, but in the long-run results in PLIN3 + LD consumption, which may have consequences on the astrocytes' MHCII distribution capacity. Taken together, our findings present a novel function of PLIN3 + LDs that can be of relevance for AD and other inflammatory conditions.
Collapse
Affiliation(s)
- Chiara Beretta
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, Uppsala, SE-752 37, Sweden
| | - Abdulkhalek Dakhel
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, Uppsala, SE-752 37, Sweden
| | - Khalid Eltom
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, Uppsala, SE-752 37, Sweden
| | - Fredrik Rosqvist
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, BMC, Uppsala University, Uppsala, Sweden
- Department of Food Studies, Nutrition and Dietetics, BMC, Uppsala University, Uppsala, SE-751 23, Sweden
| | - Simon Uzoni
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, 41390, Sweden
| | - Tobias Mothes
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, Uppsala, SE-752 37, Sweden
| | - John S Fletcher
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, 41390, Sweden
| | - Ulf Risérus
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, BMC, Uppsala University, Uppsala, Sweden
| | - Dag Sehlin
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, Uppsala, SE-752 37, Sweden
| | - Jinar Rostami
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, Uppsala, SE-752 37, Sweden
| | - Wojciech Piotr Michno
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, Uppsala, SE-752 37, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, SE-752 37, Sweden
| | - Anna Erlandsson
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, Uppsala, SE-752 37, Sweden.
| |
Collapse
|
25
|
Kopalli SR, Behl T, Baldaniya L, Ballal S, Joshi KK, Arya R, Chaturvedi B, Chauhan AS, Verma R, Patel M, Jain SK, Wal A, Gulati M, Koppula S. Neuroadaptation in neurodegenerative diseases: compensatory mechanisms and therapeutic approaches. Prog Neuropsychopharmacol Biol Psychiatry 2025; 139:111375. [PMID: 40280271 DOI: 10.1016/j.pnpbp.2025.111375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
Progressive neuronal loss is a hallmark of neurodegenerative diseases including Alzheimer's, Parkinson's, Huntington's, and Amyotrophic Lateral Sclerosis (ALS), which cause cognitive and motor impairment. Delaying the onset and course of symptoms is largely dependent on neuroadaptation, the brain's ability to restructure in response to damage. The molecular, cellular, and systemic processes that underlie neuroadaptation are examined in this study. These mechanisms include gliosis, neurogenesis, synaptic plasticity, and changes in neurotrophic factors. Axonal sprouting, dendritic remodelling, and compensatory alterations in neurotransmitter systems are important adaptations observed in NDDs; nevertheless, these processes may shift to maladaptive plasticity, which would aid in the advancement of the illness. Amyloid and tau pathology-induced synaptic alterations in Alzheimer's disease emphasize compensatory network reconfiguration. Dopamine depletion causes a major remodelling of the basal ganglia in Parkinson's disease, and non-dopaminergic systems compensate. Both ALS and Huntington's disease rely on motor circuit rearrangement and transcriptional dysregulation to slow down functional deterioration. Neuroadaptation is, however, constrained by oxidative stress, compromised autophagy, and neuroinflammation, particularly in elderly populations. The goal of emerging therapy strategies is to improve neuroadaptation by pharmacologically modifying neurotrophic factors, neuroinflammation, and synaptic plasticity. Neurostimulation, cognitive training, and physical rehabilitation are instances of non-pharmacological therapies that support neuroplasticity. Restoring compensating systems may be possible with the use of stem cell techniques and new gene treatments. The goal of future research is to combine biomarkers and individualized medicines to maximize neuroadaptive responses and decrease the course of illness. In order to reduce neurodegeneration and enhance patient outcomes, this review highlights the dual function of neuroadaptation in NDDs and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Punjab-140306, India
| | - Lalji Baldaniya
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot 360003, Gujarat, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, India; Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Renu Arya
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - Bhumi Chaturvedi
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Ashish Singh Chauhan
- Uttaranchal Institute of Pharmaceutical Sciences, Division of research and innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Rakesh Verma
- Department of Pharmacology, Institute of Medical Science, BHU, Varanasi, India
| | - Minesh Patel
- Department of Pharmacology & Pharmacy Practice, Saraswati Institute of Pharmaceutical Sciences, Dhanap, Gandhinagar, Gujarat, India
| | - Sanmati Kumar Jain
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Koni, Bilaspur, India, 495009
| | - Ankita Wal
- Pranveer Singh Institute of Technology, Pharmacy, NH-19, Bhauti Road, Kanpur, UP, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India; ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| |
Collapse
|
26
|
Wei S, Li C, Li W, Yuan F, Kong J, Su X, Huang P, Guo H, Xu J, Sun H. Glial changes and gene expression in Alzheimer's disease from snRNA-Seq and spatial transcriptomics. J Alzheimers Dis 2025:13872877251330320. [PMID: 40267277 DOI: 10.1177/13872877251330320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
BackgroundAlzheimer's disease (AD) is characterized by cortical atrophy, glutamatergic neuron loss, and cognitive decline. However, large-scale quantitative assessments of cellular changes during AD pathology remain scarce.ObjectiveThis study aims to integrate single-nuclei sequencing data from the Seattle Alzheimer's Disease Cortical Atlas (SEA-AD) with spatial transcriptomics to quantify cellular changes in the prefrontal cortex and temporal gyrus, regions vulnerable to AD neuropathological changes (ADNC).MethodsWe mapped differentially expressed genes (DEGs) and analyzed their interactions with pathological factors such as APOE expression and Lewy bodies. Cellular proportions were assessed, focusing on neurons, glial cells, and immune cells.ResultsRORB-expressing L4-like neurons, though vulnerable to ADNC, exhibited stable cell numbers throughout disease progression. In contrast, astrocytes displayed increased reactivity, with upregulated cytokine signaling and oxidative stress responses, suggesting a role in neuroinflammation. A reduction in synaptic maintenance pathways indicated a decline in astrocytic support functions. Microglia showed heightened immune surveillance and phagocytic activity, indicating their role in maintaining cortical homeostasis.ConclusionsThe study underscores the critical roles of glial cells, particularly astrocytes and microglia, in AD progression. These findings contribute to a better understanding of cellular dynamics and may inform therapeutic strategies targeting glial cell function in AD.
Collapse
Affiliation(s)
- Songren Wei
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Institute for Brain Science and Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| | - Chenyang Li
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Institute for Brain Science and Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | | | - Fumiao Yuan
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jingjing Kong
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xi Su
- Women and Children Medical Research Center, Affiliated Foshan Women and Children Hospital, Foshan, China
| | - Peng Huang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Women and Children Medical Research Center, Affiliated Foshan Women and Children Hospital, Foshan, China
| | - Hongbo Guo
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Institute for Brain Science and Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiangping Xu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| | - Haitao Sun
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Institute for Brain Science and Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
27
|
Lemes JA, Rosário BDA, Rocha SMS, Bandeira SS, Ribeiro AM, Vaz SH, Sebastião AM, Armada-Moreira A, Ribeiro DA, de Barros Viana M. The role of glutamate receptors and transporters in epilepsy: evidence from animal studies. Rev Neurosci 2025:revneuro-2024-0173. [PMID: 40248882 DOI: 10.1515/revneuro-2024-0173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/06/2025] [Indexed: 04/19/2025]
Abstract
Epilepsy encompasses a group of chronic brain disorders characterized by recurrent, hypersynchronous activity of neuronal clusters, with epileptic seizures being the primary manifestation of these disorders. The objective of epilepsy treatment is to prevent seizures with minimum adverse side effects. However, approximately 30 % of patients do not respond to available medications. One proposed mechanism of epileptogenesis is glutamate excitotoxicity. When released in excess or not appropriately removed from the synaptic cleft, glutamate hyperactivates receptors, causing a biochemical cascade, which culminates in seizures and cell death. The use of animal models is essential for uncovering potential epileptogenic pathways, understanding the role of receptors and transporters in excitotoxicity, and screening effective antiepileptic treatments. This review examines studies that investigate the role of glutamate transporters and receptors in excitotoxicity and epileptogenesis using animal models. For this, we searched through both PubMed/Medline and ScienceDirect databases. After applying the inclusion and exclusion criteria, 26 (twenty-six) studies were selected for analysis. The studies addressed key glutamate transporter family of excitatory amino acid transporters (EAATs) EAAT1, EAAT2, and EAAT3, responsible for glutamate clearance, as well as AMPA receptor subunits GluA1 and GluA2, NMDA receptor subunits GluN1, GluN2a, and GluN2b, and the metabotropic receptors mGluR5 and mGluR2/3. Results showed that the dysregulation of these transporters and receptors is associated to seizure induction and excitotoxic damage, pointing to their fundamental role in the mechanisms of excitotoxicity and epileptogenesis. These findings highlight the potential of targeting glutamate transporters and receptors to stabilize glutamate homeostasis as an intervention in epilepsy management.
Collapse
Affiliation(s)
- Jéssica Alves Lemes
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Rua Silva Jardim, 136, 11015-021, Santos, SP, Brazil
| | - Barbara Dos Anjos Rosário
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Rua Silva Jardim, 136, 11015-021, Santos, SP, Brazil
| | - Sophia Morya Santos Rocha
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Rua Silva Jardim, 136, 11015-021, Santos, SP, Brazil
| | - Susana Sieiro Bandeira
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Rua Silva Jardim, 136, 11015-021, Santos, SP, Brazil
| | - Alessandra Mussi Ribeiro
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Rua Silva Jardim, 136, 11015-021, Santos, SP, Brazil
| | - Sandra Henriques Vaz
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
- Gulbenkian Institute for Molecular Medicine, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
- Centro Cardiovascular da Universidade de Lisboa, CCUL (CCUL@RISE), Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Ana Maria Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
- Gulbenkian Institute for Molecular Medicine, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
- Centro Cardiovascular da Universidade de Lisboa, CCUL (CCUL@RISE), Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Adam Armada-Moreira
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125, Modena, Italy
| | - Daniel Araki Ribeiro
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Rua Silva Jardim, 136, 11015-021, Santos, SP, Brazil
| | - Milena de Barros Viana
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Rua Silva Jardim, 136, 11015-021, Santos, SP, Brazil
| |
Collapse
|
28
|
Mosora O, Maier S, Manu D, Bărcuțean L, Roman M, Dumitreasă M, Bălașa R. Exosomal microRNAs as Early Transition Biomarkers from Recurrent-Remissive to Secondary Progressive Multiple Sclerosis. Int J Mol Sci 2025; 26:3889. [PMID: 40332781 PMCID: PMC12028311 DOI: 10.3390/ijms26083889] [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: 03/02/2025] [Revised: 04/17/2025] [Accepted: 04/19/2025] [Indexed: 05/08/2025] Open
Abstract
Multiple sclerosis (MS) is a chronic, immune-mediated disease that affects young adults, leading to neurological disability. Regardless of the studies and the research involved in developing an efficient disease-modifying therapy (DMT), relapsing-remitting multiple sclerosis (RRMS) will transition to a progressive multiple sclerosis phenotype. The moment of transition from RRMS to secondary progressive multiple sclerosis (SPMS) is difficult to predict, and the diagnosis is based on the accumulation of disabilities in the evolution of the disease. Research on microRNAs' (miRNAs) role in MS began in the early 2000s, with miR-155 frequently cited for its link to blood-brain barrier dysfunction and neurodegeneration, making it an early transition biomarker from RRMS to SPMS. The purpose of this review is to reveal the importance of finding a biomarker from the molecular field that will be able to identify the transition phase so patients can receive high-efficacy treatments and to cease the clinical progression.
Collapse
Affiliation(s)
- Oana Mosora
- Doctoral School, “George Emil Palade” University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (O.M.); (R.B.)
- Ist Neurology Clinical, Emergency Clinical County Hospital Targu Mures, 540136 Targu Mures, Romania; (L.B.); (M.R.); (M.D.)
| | - Smaranda Maier
- Ist Neurology Clinical, Emergency Clinical County Hospital Targu Mures, 540136 Targu Mures, Romania; (L.B.); (M.R.); (M.D.)
- Department of Neurology, “George Emil Palade” University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540136 Targu Mures, Romania
| | - Doina Manu
- Center for Advanced Medical and Pharmaceutical Research, “George Emil Palade” University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Laura Bărcuțean
- Ist Neurology Clinical, Emergency Clinical County Hospital Targu Mures, 540136 Targu Mures, Romania; (L.B.); (M.R.); (M.D.)
- Department of Neurology, “George Emil Palade” University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540136 Targu Mures, Romania
| | - Medeea Roman
- Ist Neurology Clinical, Emergency Clinical County Hospital Targu Mures, 540136 Targu Mures, Romania; (L.B.); (M.R.); (M.D.)
| | - Mihai Dumitreasă
- Ist Neurology Clinical, Emergency Clinical County Hospital Targu Mures, 540136 Targu Mures, Romania; (L.B.); (M.R.); (M.D.)
| | - Rodica Bălașa
- Doctoral School, “George Emil Palade” University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (O.M.); (R.B.)
- Ist Neurology Clinical, Emergency Clinical County Hospital Targu Mures, 540136 Targu Mures, Romania; (L.B.); (M.R.); (M.D.)
- Department of Neurology, “George Emil Palade” University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540136 Targu Mures, Romania
| |
Collapse
|
29
|
Zhao X, Zhang S, Wu M, Zhang B, Wan G, Zhang M, Li J, Fei Z, Zhu G, Jiang S, Xiao M, Liu W, Zhao Z, Huang B, Ran J. High urea promotes mitochondrial fission and functional impairments in astrocytes inducing anxiety-like behavior in chronic kidney disease mice. Metab Brain Dis 2025; 40:186. [PMID: 40244426 DOI: 10.1007/s11011-025-01612-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 04/11/2025] [Indexed: 04/18/2025]
Abstract
High urea can induce depression and anxiety. Activation of astrocytes is closely associated with psychiatric disorders. However, the pathological mechanism of whether high urea affects astrocyte structure and function to induce anxiety-like behaviors remain unclear. We established a high-urea chronic kidney disease (CKD) mouse model and found that these mice exhibited elevated levels of anxiety through behavioral experiments. Immunofluorescence and transmission electron microscopy studies of astrocytes revealed a decrease in density and branching of mPFC astrocytes. Additionally, we observed a significant reduction in ATP and BDNF levels in the mPFC and primary astrocytes of CKD mice induced by high urea. Analysis of gene expression differences in astrocytes between WT and high-urea mice indicated alterations in mitochondrial dynamics-related signaling pathways in astrocytes. We established a high-urea primary astrocyte model to assess mitochondrial function and levels of fusion and fission proteins. Treatment of primary astrocytes with high urea led to mitochondrial fragmentation and downregulation of Mfn2 expression. These results suggested that high urea downregulates Mfn2 expression in mPFC astrocytes, induced mitochondrial fusion-fission abnormalities, disrupted astrocyte energy metabolism, and promoted high-urea-related anxiety. Mfn2 may represent a potential therapeutic target for high-urea-related anxiety.
Collapse
Affiliation(s)
- Xi Zhao
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Shengyao Zhang
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Mengna Wu
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Binyun Zhang
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Guoran Wan
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Meng Zhang
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Jing Li
- Department of Stem Cell and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Zhuo Fei
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Guoqi Zhu
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Shaoqiu Jiang
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Mohan Xiao
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Wanjia Liu
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Zhelun Zhao
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Boyue Huang
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China.
| | - Jianhua Ran
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China.
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China.
- Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
30
|
Vijayaraghavan M, Murali SP, Thakur G, Li XJ. Role of glial cells in motor neuron degeneration in hereditary spastic paraplegias. Front Cell Neurosci 2025; 19:1553658. [PMID: 40302786 PMCID: PMC12037628 DOI: 10.3389/fncel.2025.1553658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
This review provides a comprehensive overview of hereditary spastic paraplegias (HSPs) and summarizes the recent progress on the role of glial cells in the pathogenesis of HSPs. HSPs are a heterogeneous group of neurogenetic diseases characterized by axonal degeneration of cortical motor neurons, leading to muscle weakness and atrophy. Though the contribution of glial cells, especially astrocytes, to the progression of other motor neuron diseases like amyotrophic lateral sclerosis (ALS) is well documented, the role of glial cells and the interaction between neurons and astrocytes in HSP remained unknown until recently. Using human pluripotent stem cell-based models of HSPs, a study reported impaired lipid metabolisms and reduced size of lipid droplets in HSP astrocytes. Moreover, targeting lipid dysfunction in astrocytes rescues axonal degeneration of HSP cortical neurons, demonstrating a non-cell-autonomous mechanism in axonal deficits of HSP neurons. In addition to astrocytes, recent studies revealed dysfunctions in HSP patient pluripotent stem cell-derived microglial cells. Increased microgliosis and pro-inflammation factors were also observed in HSP patients' samples, pointing to an exciting role of innate immunity and microglia in HSP. Building upon these recent studies, further investigation of the detailed molecular mechanism and the interplay between glial cell dysfunction and neuronal degeneration in HSP by combining human stem cell models, animal models, and patient samples will open avenues for identifying new therapeutic targets and strategies for HSP.
Collapse
Affiliation(s)
- Manaswini Vijayaraghavan
- Department of Biomedical Sciences, University of Illinois College of Medicine Rockford, Rockford, IL, United States
| | - Sarvika Periyapalayam Murali
- Department of Biomedical Sciences, University of Illinois College of Medicine Rockford, Rockford, IL, United States
| | - Gitika Thakur
- Department of Biomedical Sciences, University of Illinois College of Medicine Rockford, Rockford, IL, United States
| | - Xue-Jun Li
- Department of Biomedical Sciences, University of Illinois College of Medicine Rockford, Rockford, IL, United States
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
31
|
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
|
32
|
Nentwig TB, Obray JD, Kruyer A, Wilkes ET, Vaughan DT, Scofield MD, Chandler LJ. Central amygdala astrocyte plasticity underlies GABAergic dysregulation in ethanol dependence. Transl Psychiatry 2025; 15:132. [PMID: 40199844 PMCID: PMC11978928 DOI: 10.1038/s41398-025-03337-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/21/2025] [Accepted: 03/19/2025] [Indexed: 04/10/2025] Open
Abstract
Dependence is a hallmark of alcohol use disorder characterized by excessive alcohol intake and withdrawal symptoms. The central nucleus of the amygdala (CeA) is a key brain structure underlying the synaptic and behavioral consequences of ethanol dependence. While accumulating evidence suggests that astrocytes regulate synaptic transmission and behavior, there is a limited understanding of the role astrocytes play in ethanol dependence. The present study used a combination of viral labeling, super resolution confocal microscopy, 3D image analysis, and slice electrophysiology to determine the effects of chronic intermittent ethanol (CIE) exposure on astrocyte plasticity in the CeA. During withdrawal from CIE exposure, we observed increased GABA transmission, an upregulation in astrocytic GAT3 levels, and an increased proximity of astrocyte processes near CeA synapses. Furthermore, GAT3 levels and synaptic proximity were positively associated with voluntary ethanol drinking in dependent rats. Slice electrophysiology confirmed that the upregulation in astrocytic GAT3 levels was functional, as CIE exposure unmasked a GAT3-sensitive tonic GABA current in the CeA. A causal role for astrocytic GAT3 in ethanol dependence was assessed using viral-mediated GAT3 overexpression and knockdown approaches. However, GAT3 knockdown or overexpression had no effect on somatic withdrawal symptoms, dependence-escalated ethanol intake, aversion-resistant drinking, or post-dependent ethanol drinking in male or female rats. Moreover, intra-CeA pharmacological inhibition of GAT3 did not alter dependent ethanol drinking. Together, these findings indicate that ethanol dependence induces GABAergic dysregulation and astrocyte plasticity in the CeA. However, these changes in astrocytic GAT3 do not appear to be necessary for the drinking related phenotypes associated with dependence.
Collapse
Affiliation(s)
- Todd B Nentwig
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - J Daniel Obray
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Anna Kruyer
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
- Department of Neuroscience, University of Cincinnati, Cincinnati, OH, USA
| | - Erik T Wilkes
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Dylan T Vaughan
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael D Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - L Judson Chandler
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
33
|
Liu ZH, Chen NY, Huang CY, Lin YJ, Yip PK, Wei KC, Liu HL. Modulation of the immune response by focused ultrasound suppressed brain abscess formation. Drug Deliv Transl Res 2025:10.1007/s13346-025-01847-3. [PMID: 40193008 DOI: 10.1007/s13346-025-01847-3] [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] [Accepted: 03/23/2025] [Indexed: 04/12/2025]
Abstract
Brain abscess is a serious, life-threatening intracranial infection caused by inflammation and collection of infected material. Given the rise of multi-drug resistant strains and the widespread presence of bacteria, it is probable that the incidence of brain abscesses is expected to endure. The sequela of brain abscess constitutes a major source of morbidity and mortality. Brain abscess may cause permanent neurological damage, such as paresis, hydrocephalus, spasticity, mental deterioration and epileptic seizure. Current therapeutic approaches include surgical excision or drainage combined with prolonged antimicrobial treatment usually lasting 6-8 weeks. However, extended antimicrobial treatment may cause adverse side effects, such as nephrotoxicity, ototoxicity, and bone marrow suppression. As a result, it is essential to develop a novel approach to facilitate antibiotics delivery and shorten the therapeutic course clinically. Recently, focused ultrasound (FUS) has been demonstrated to have an ability to temporally open the brain blood barrier (BBB) and modulate the immune response in the brain tumor animal model or in naïve animals. In our study, we demonstrated the focused ultrasound treatment (3W acoustic power, 0.6 MPa peak negative pressure) to treat brain abscess by boosting immune response in CNS infection in the brain abscess animal model. The size of the brain abscess is reduced by 50 percent when the MRI scan is taken at 3 weeks post-treatment. The animals get better recovery after treatment. The use of low intensity FUS with systemic microbubble infusion to open the BBB by mechanical acoustic cavitation elicited an immediate immune response including elevations in proinflmmatory cytokine (IL-1, TNFα and IL-6) in the brain parenchyma surround the brain abscess. Furthermore, FUS exposure treatment also activated glial cells, potentially enhancing the encapsulation of brain abscesses and reducing the spread of bacteria to the adjacent brain parenchyma. Histological analysis also demonstrated that FUS can reduce neuron loss and blood vessel damage during brain abscess formation. Our findings indicate that the FUS system can achieve local reversible BBB opening, enhancing immunomodulation in an animal model of brain abscess.
Collapse
Affiliation(s)
- Zhuo-Hao Liu
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Chang Gung Medical College and University, Taoyuan County, 33305, Taiwan
| | - Nan-Yu Chen
- Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Chang Gung Medical College and University, Taoyuan County, 33305, Taiwan
| | - Chiung-Yin Huang
- Neuroscience Research Center, Chang Gung Memorial Hospital, Taoyuan, 33305, Taiwan
| | - Ya-Jui Lin
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Chang Gung Medical College and University, Taoyuan County, 33305, Taiwan
| | - Ping K Yip
- Barts and the London School of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, E1 2AT, UK
| | - Kuo-Chen Wei
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Chang Gung Medical College and University, Taoyuan County, 33305, Taiwan.
- Neuroscience Research Center, Chang Gung Memorial Hospital, Taoyuan, 33305, Taiwan.
| | - Hao-Li Liu
- Department of Electrical Engineering, National Taiwan University, Taipei, 10617, Taiwan.
| |
Collapse
|
34
|
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] [Download PDF] [Figures] [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
|
35
|
Colombari E, Biancardi VC, Colombari DSA, Katayama PL, Medeiros FDCD, Aitken AV, Xavier CH, Pedrino GR, Bragin DE. Hypertension, blood-brain barrier disruption and changes in intracranial pressure. J Physiol 2025; 603:2245-2261. [PMID: 40163552 DOI: 10.1113/jp285058] [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: 08/30/2024] [Accepted: 03/10/2025] [Indexed: 04/02/2025] Open
Abstract
Intracranial pressure (ICP) is pressure within the cranium, between 5 and 15 mmHg in a normal brain, and is influenced by the dynamic balance between brain tissue, cerebrospinal fluid (CSF) and cerebral blood volume. ICP is vital for cerebral health, impacting outcomes in various neurological conditions. Disruptions, such as cerebral haemorrhage, hydrocephalus and malignant hypertension, can lead to elevated ICP, a dangerous condition known as intracranial hypertension (IH). Systemic hypertension significantly impacts cerebral health by causing microvascular damage, dysfunction of the blood-brain barrier (BBB) and impairment of intracranial compliance (ICC). This increases the risk of IH), cerebral ischaemia, neuroinflammation and lacunar infarction, further worsening neurological dysfunction. This review describes the complex relationship between hypertension and ICP regulation, focusing on the mechanisms underlying ICP and ICC adjustments in hypertensive conditions and emphasizing the role of BBB integrity and cerebral blood flow (CBF) dynamics. It discusses how the sympathetic output might change the regulation of CBF and the maintenance of ICP, highlighting how hypertensive conditions can impair this mechanism, increasing the risk of cerebral ischaemia. The neurovascular unit, including astrocytes and microglia, plays a significant role in this process, contributing to IH in hypertensive patients. Understanding the effects of hypertension on ICP and ICC could lead to therapies aimed at preserving BBB integrity, reducing inflammation and improving cerebral compliance, potentially preventing brain dysfunction and reducing stroke risk in hypertensive patients. This review underscores the need for early detection and intervention to mitigate the severe consequences of uncontrolled hypertension on cerebral health.
Collapse
Affiliation(s)
- Eduardo Colombari
- Department of Physiology and Pathology, School of Dentistry of Araraquara, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Vinícia Campana Biancardi
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Débora Simões Almeida Colombari
- Department of Physiology and Pathology, School of Dentistry of Araraquara, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Pedro Lourenço Katayama
- Department of Physiology and Pathology, School of Dentistry of Araraquara, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Fernanda de Campos de Medeiros
- Department of Physiology and Pathology, School of Dentistry of Araraquara, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Andrew Vieira Aitken
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Carlos Henrique Xavier
- Department of Physiological Science, Biological Science Institute, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Gustavo Rodrigues Pedrino
- Department of Physiological Science, Biological Science Institute, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Denis E Bragin
- Lovelace Biomedical Research Institute, Albuquerque, New Mexico, USA
- Department of Neurology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| |
Collapse
|
36
|
Talifu Z, Xu X, Du H, Li Z, Wang X, Zhang C, Pan Y, Ke H, Liu W, Gao F, Yang D, Jing Y, Yu Y, Du L, Li J. Effect of in vivo reprogramming of astrocytes combined with exercise training on neurorepair in rats with spinal cord injury. Animal Model Exp Med 2025; 8:595-605. [PMID: 39844772 PMCID: PMC12008439 DOI: 10.1002/ame2.12545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 12/15/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND The inability of damaged neurons to regenerate and of axons to establish new functional connections leads to permanent functional deficits after spinal cord injury (SCI). Although astrocyte reprogramming holds promise for neurorepair in various disease models, it is not sufficient on its own to achieve significant functional recovery. METHODS A rat SCI model was established using a spinal cord impactor. Seven days postsurgery, adeno-associated virus were injected to overexpress the transcription factors NeuroD1 and Neurogenin-2 (Ngn2) in the spinal cord. The rats were then trained to walk on a weight-supported treadmill for 4 weeks, starting 14 days after modeling. The effects of these interventions on motor and sensory functions, as well as spinal cord tissue repair, were subsequently evaluated. RESULTS The combination of NeuroD1 and Ngn2 overexpression with weight-supported exercise training significantly improved gait compared to either intervention alone. The group receiving the combined intervention exhibited enhanced sensitivity in sensory assessments. Immunofluorescence analysis revealed increased colocalization of astrocytes and microtubule-associated protein 2-positive neurons in the injury area. These effects were more pronounced than those observed with spinal cord tissue repair alone. Additionally, the combined intervention significantly reduced glial scarring and the size of the injury area. CONCLUSION Exercise intervention enhances the reprogramming effects of astrocytes and restores motor function, yielding better results than either intervention alone.
Collapse
Affiliation(s)
- Zuliyaer Talifu
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Chinese Institute of Rehabilitation ScienceBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
- School of Population Medicine and Public HealthChinese Academy of Medical Sciences/Peking Union Medical CollegeBeijingChina
| | - Xin Xu
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Neurology, Cheeloo College of MedicineShandong University, Qilu Hospital of Shandong UniversityJinanChina
- School of Health and Life SciencesUniversity of Health and Rehabilitation SciencesQingdaoChina
| | - Huayong Du
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Chinese Institute of Rehabilitation ScienceBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
| | - Zehui Li
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Chinese Institute of Rehabilitation ScienceBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
| | - Xiaoxin Wang
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Chinese Institute of Rehabilitation ScienceBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
| | - Chunjia Zhang
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Chinese Institute of Rehabilitation ScienceBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
| | - Yunzhu Pan
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Chinese Institute of Rehabilitation ScienceBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
| | - Han Ke
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Department of Neurology, Cheeloo College of MedicineShandong University, Qilu Hospital of Shandong UniversityJinanChina
- School of Health and Life SciencesUniversity of Health and Rehabilitation SciencesQingdaoChina
| | - Wubo Liu
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Department of Neurology, Cheeloo College of MedicineShandong University, Qilu Hospital of Shandong UniversityJinanChina
- School of Health and Life SciencesUniversity of Health and Rehabilitation SciencesQingdaoChina
| | - Feng Gao
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
| | - Degang Yang
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
| | - Yingli Jing
- School of RehabilitationCapital Medical UniversityBeijingChina
- Chinese Institute of Rehabilitation ScienceBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
| | - Yan Yu
- School of RehabilitationCapital Medical UniversityBeijingChina
- Chinese Institute of Rehabilitation ScienceBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
| | - Liangjie Du
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
| | - Jianjun Li
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Chinese Institute of Rehabilitation ScienceBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
- School of Health and Life SciencesUniversity of Health and Rehabilitation SciencesQingdaoChina
| |
Collapse
|
37
|
Thaele A, Barba L, Abu-Rumeileh S, Foschi M, Otto M. Neurofilament light chain and glial fibrillary acidic protein as diagnostic and prognostic biomarkers in epileptic seizures and epilepsy: A systematic review. Epilepsy Behav 2025; 165:110321. [PMID: 39983592 DOI: 10.1016/j.yebeh.2025.110321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/25/2025] [Accepted: 02/11/2025] [Indexed: 02/23/2025]
Abstract
Epileptology - with epilepsy as one of the most common neurological diseases - has an urgent need for easily accessible biomarkers to improve diagnosis, prognosis and therapeutic monitoring. Neurofilament light chain (NfL) and Glial Fibrillary Acidic Protein (GFAP) have emerged as promising fluid biomarkers in various neurological disorders. Their potential role in epileptic seizures and epilepsy remains largely unexplored. To assess the current state of research on this topic we comprehensively searched the published literature for studies on GFAP and/or NfL in cerebrospinal fluid and/or blood in adult humans with epileptic seizures, status epilepticus or epilepsy (last data base search on 10th of May 2024). We identified a total of 2285 publications of which 19 fulfilled our search criteria. The studies targeted various outcomes such as prognosis in status epilepticus, differentiation of seizure semiology and etiology, differentiation of epileptic seizures from non-epileptic conditions, prediction of epilepsy in autoimmune epilepsy, after a stroke or after a first unprovoked seizure, the role of the time interval from seizure to sampling, the association with disease duration as well as seizure frequency and the influence of seizure suppressing medication. The results are heterogeneous but indicate promising applications for both NfL and GFAP in diagnosis and prognostication of patients with epileptic seizures and epilepsy. In the present review we summarize the current evidence, future perspectives, but also limitations, of NfL and GFAP as fluid biomarkers in epilepsy and epileptic seizures.
Collapse
Affiliation(s)
- Annemarie Thaele
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany.
| | - Lorenzo Barba
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany
| | - Samir Abu-Rumeileh
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany
| | - Matteo Foschi
- Department of Neuroscience, S. Maria delle Croci Hospital, AUSL Romagna, Ravenna, Italy; Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Markus Otto
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
38
|
Ziar R, Tesar PJ, Clayton BLL. Astrocyte and oligodendrocyte pathology in Alzheimer's disease. Neurotherapeutics 2025; 22:e00540. [PMID: 39939240 PMCID: PMC12047399 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
|
39
|
Fu Y, Adler GL, Youssef P, Phan K, Halliday GM, Dzamko N, Kim WS. Human Endogenous Retrovirus K in Astrocytes Is Altered in Parkinson's Disease. Mov Disord 2025; 40:683-692. [PMID: 39840837 PMCID: PMC12006878 DOI: 10.1002/mds.30128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/01/2025] [Accepted: 01/06/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Parkinson's disease (PD) is the most common neurodegenerative movement disease. Human endogenous retroviruses (HERVs) are proviral remnants of ancient retroviral infection of germ cells that now constitute about 8% of the human genome. Under certain disease conditions, HERV genes are activated and partake in the disease process. However, virtually nothing is known about the pathological relationship, if any, between HERV and PD. OBJECTIVE The objectives of this study were to unravel the pathological relationship between human endogenous retrovirus K (HERV-K) and PD, determine the localization of HERV-K in the brain, determine whether HERV-K levels are altered in PD brain and blood, and examine whether HERV-K could serve as a biomarker for PD. METHODS In situ HERV-K and glial fibrillary acidic protein (GFAP) expression in the superior frontal and fusiform cortices of PD and control brain were analyzed using immunofluorescence and confocal microscopy. HERV-K load and copy number in PD and control blood were measured by digital droplet polymerase chain reaction and GFAP by single-molecule array. HERV-K load was analyzed in relation to the Hoehn and Yahr Scale and Movement Disorder Society Unified Parkinson's Disease Rating Scale Part III. RESULTS HERV-K is predominantly expressed in astrocytes and colocalized with astrocytic GFAP, with decreased expression of both HERV-K and GFAP in PD brain compared with controls. Consistent with this, HERV-K levels were decreased in PD blood compared with controls and were correlated to blood GFAP levels. HERV-K levels were inversely correlated to PD severity and duration. CONCLUSIONS These findings suggest that HERV-K is related to astrocyte function and to PD progression, and that HERV-K could be neuroprotective. © 2025 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- YuHong Fu
- Brain and Mind CentreThe University of SydneySydneyNew South WalesAustralia
- School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
| | - Gabrielle L. Adler
- Brain and Mind CentreThe University of SydneySydneyNew South WalesAustralia
- School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
| | - Priscilla Youssef
- Brain and Mind CentreThe University of SydneySydneyNew South WalesAustralia
- School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
| | - Katherine Phan
- Brain and Mind CentreThe University of SydneySydneyNew South WalesAustralia
- School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
| | - Glenda M. Halliday
- Brain and Mind CentreThe University of SydneySydneyNew South WalesAustralia
- School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
| | - Nicolas Dzamko
- Brain and Mind CentreThe University of SydneySydneyNew South WalesAustralia
- School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
| | - Woojin Scott Kim
- Brain and Mind CentreThe University of SydneySydneyNew South WalesAustralia
- School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
| |
Collapse
|
40
|
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
|
41
|
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
|
42
|
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
|
43
|
Huang S, Lin L, Gao X, Li Y, Qiu H, Deng C, Qian L, Chen Y, Tang W, Liang Y, Su S, Yang Z. Glymphatic system dysfunction in pediatric tourette syndrome Neuroimaging evidence from MRI metrics. J Psychiatr Res 2025; 184:1-7. [PMID: 40031125 DOI: 10.1016/j.jpsychires.2025.02.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND AND OBJECTIVES The glymphatic system, vital for brain waste clearance, is implicated in neurodevelopmental diseases, but its role in pediatric Tourette syndrome (TS) is not well understood. This study investigates structural and functional alterations in the glymphatic system in pediatric TS using non-invasive MRI techniques. METHODS This case-control study included 37 children with Tourette syndrome (TS) and 37 age- and gender-matched typically developing (TD) controls. We assessed brain volumetric differences and glymphatic function using two MRI metrics: perivascular space (PVS) burden for glymphatic influx and the DTI-ALPS index for waste clearance, with PVS quantified via semi-automated analysis of axial T2-weighted images. Correlations between MRI metrics and clinical symptoms in TS children were analyzed using partial correlations. RESULTS Children with Tourette syndrome (TS) exhibited significant reductions in brain parenchymal and white matter volume compared to typically developing (TD) children (all PFDR < 0.001), along with a higher perivascular space (PVS) volume (6.29 ± 3.62 mL vs. 4.76 ± 2.13 mL; PFDR = 0.046), indicating impaired glymphatic influx. The DTI-ALPS index was lower in TS (1.21 ± 0.18 vs. 1.46 ± 0.12; PFDR < 0.001), reflecting reduced waste clearance, and Dzassoc and Dzproj metrics were positively correlated with motor tic severity in TS (all P ≤ 0.02). CONCLUSIONS Our findings suggest significant glymphatic dysfunction in pediatric TS, indicating its role in the disorder's pathogenesis. Increased PVS burden and decreased DTI-ALPS index may serve as non-invasive biomarkers for diagnosing and understanding TS mechanisms.
Collapse
Affiliation(s)
- Shuzhen Huang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Liping Lin
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiang Gao
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yufen Li
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Huaqiong Qiu
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Chengfen Deng
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Long Qian
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 100817, China
| | - Yingqian Chen
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Wen Tang
- Department of Pediatric Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yujian Liang
- Department of Pediatric Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shu Su
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhiyun Yang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
44
|
Krause S, Florea A, Choi CH, Worthoff WA, Heinzel A, Fischer S, Burda N, Neumaier B, Shah NJ, Lohmann P, Mottaghy FM, Langen KJ, Stegmayr C. Autoradiography of Intracerebral Tumours in the Chick Embryo Model: A Feasibility Study Using Different PET Tracers. Mol Imaging Biol 2025; 27:151-162. [PMID: 39838234 PMCID: PMC12062108 DOI: 10.1007/s11307-025-01983-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/19/2024] [Accepted: 01/03/2025] [Indexed: 01/23/2025]
Abstract
PURPOSE In addition to rodent models, the chick embryo model has gained attention for radiotracer evaluation. Previous studies have investigated tumours on the chorioallantoic membrane (CAM), but its value for radiotracer imaging of intracerebral tumours has yet to be demonstrated. PROCEDURES Human U87 glioblastoma cells and U87-IDH1 mutant glioma cells were implanted into the brains of chick embryos at developmental day 5. After 12-14 days of tumour growth, blood-brain-barrier integrity was evaluated in vivo using MRI contrast enhancement or ex vivo with Evans blue dye. The tracers O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) (n = 5), 3,4-dihydroxy-6-[18F]-fluoro-L-phenylalanine ([18F]FDOPA) (n = 3), or [68Ga] labelled quinoline-based small molecule fibroblast activation protein inhibitor ([68Ga]FAPI-46) (n = 4) were injected intravenously if solid tumours were detected with MRI. For time-activity curves for [18F]FET, additional micro PET (µPET) was performed. The chick embryos were sacrificed 60 min post-injection, and cryosections of the tumour-bearing brains were produced and evaluated with autoradiography and immunohistochemistry. RESULTS Intracerebral tumours were produced with a 100% success rate in viable chick embryos at the experimental endpoint. However, 52% of chick embryos (n = 85) did not survive the procedure to embryonic development day 20. For the evaluated radiotracers, the tumour-to-brain ratios (TBR) derived from ex vivo autoradiography, as well as the tracer kinetics derived from µPET for intracerebral chick embryo tumours, were comparable to those previously reported in rodents and patients: the TBRmean for [18F]FET was 1.69 ± 0.54 (n = 5), and 3.8 for one hypermetabolic tumour and < 2.0 for two isometabolic tumors using [18F]FDOPA, with a TBRmean of 1.92 ± 1,11 (n = 3). The TBRmean of [68Ga]FAPI-46 for intracerebral chick embryo tumours was 19.13 ± 0.64 (n = 4). An intact blood-tumour barrier was observed in one U87-MG tumour (n = 5). CONCLUSIONS Radiotracer imaging of intracerebral tumours in the chick embryo offers a fast model for the evaluation of radiotracer uptake, accumulation, and kinetics. Our results indicate a high comparability between intracerebral tumour imaging in chick embryos and xenograft rodent models or brain tumour patients.
Collapse
Affiliation(s)
- Sandra Krause
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany.
| | - Alexandru Florea
- Department of Nuclear Medicine, RWTH Aachen University Hospital, Aachen, Germany
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Chang-Hoon Choi
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Wieland A Worthoff
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Alexander Heinzel
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
- Department for Nuclear Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Saskia Fischer
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Nicole Burda
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Bernd Neumaier
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - N Jon Shah
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
- Department of Neurology, RWTH Aachen University Hospital, Aachen, Germany
- JARA - BRAIN - Translational Medicine, Aachen, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
- Department of Nuclear Medicine, RWTH Aachen University Hospital, Aachen, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, RWTH Aachen University Hospital, Aachen, Germany
- JARA - BRAIN - Translational Medicine, Aachen, Germany
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
- Department of Nuclear Medicine, RWTH Aachen University Hospital, Aachen, Germany
| | - Carina Stegmayr
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
- Department of Nuclear Medicine, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
45
|
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
|
46
|
Mongi-Bragato B, Sánchez MA, Avalos MP, Boezio MJ, Guzman AS, Rigoni D, Perassi EM, Mas CR, Bisbal M, Bollati FA, Cancela LM. Activation of Nuclear Factor-kappa B in the nucleus accumbens core is necessary for chronic stress-induced glutamate and neuro-immune alterations that facilitate cocaine self-administration. Brain Behav Immun 2025; 128:1-15. [PMID: 40139275 DOI: 10.1016/j.bbi.2025.03.028] [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: 10/22/2024] [Revised: 03/18/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025] Open
Abstract
Stressful events are associated with impaired glutamate signaling and neuroimmune adaptations that may increase the vulnerability of individuals to cocaine addiction. We previously demonstrated that chronic stress induced reactive microglia and increased TNF-α expression in the nucleus accumbens core (NAcore), both alterations strongly linked with impaired glutamate homeostasis and the facilitation of cocaine self-administration. The nuclear factor kappa-B (NF-κB) is a critical regulator of many immune- and addiction-related genes, such as the gene coding for glutamate transporter (GLT-1), and it is considered a master regulator of inflammation, reported to be a key driver of microglia activation in psychiatric diseases. However, no studies have examined the role of NF-κB signaling within the NAcore in the neuroimmune and glutamate mechanism, underpinning stress-induced vulnerability to cocaine self-administration. Here we investigate whether viral dominant negative inhibition of I kappa B kinase (IKKdn), a signaling molecule responsible for NF-κB activation, would prevent stress-induced facilitation to cocaine self-administration and associated changes in accumbal GLT-1 and TNF-α expression. We also explore N-myc proto-oncogene protein (N-myc) levels as a link between NF-κB and stress-induced GLT-1 downregulation. For seven days (days 1-7), adult male rats were restrained for 2 h/day. Animals were administered an intra-NAcore with IKKdn or empty lentiviruses on day 14 after the first restraint stress session. Marked activation of NF-κB was detected in the NAcore of stressed subjects, along with increased NF-κB expression in astrocytes. Consistently, viral NF-κB inhibition prevented stress-induced facilitation of cocaine self-administration. Moreover, NF-κB blockade results in the restoration of stress-induced reduction in GLT-1 levels and was effective in suppressing stress-induced TNF-α within the NAcore. These findings suggest that accumbal NF-κB signaling exerts a central control over stress-altered downstream neuroimmune and glutamate function underlying vulnerability to cocaine use disorders.
Collapse
Affiliation(s)
- Bethania Mongi-Bragato
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina.
| | - Marianela Adela Sánchez
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina
| | - María Paula Avalos
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina
| | - María Julieta Boezio
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina
| | - Andrea Susana Guzman
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina
| | - Diana Rigoni
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina
| | - Eduardo Marcelo Perassi
- Instituto de Investigaciones en Físico-Química de Córdoba, INFIQC-CONICET, Departamento de Química Teórica y Computacional, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina
| | - Carlos Ruben Mas
- Centro de Investigaciones en Química Biológica de Córdoba, CIQUIBIC-CONICET, Departamento de Química Bilógica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina
| | - Mariano Bisbal
- Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET, Friuli 2434, Colinas de Vélez Sarsfield (5016) Córdoba, Argentina, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Flavia Andrea Bollati
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina.
| | - Liliana Marina Cancela
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina.
| |
Collapse
|
47
|
Gong J, Li J, Li J, He A, Ren B, Zhao M, Li K, Zhang Y, He M, Liu Y, Wang Z. Impact of Microglia-Derived Extracellular Vesicles on Resident Central Nervous System Cell Populations After Acute Brain Injury Under Various External Stimuli Conditions. Mol Neurobiol 2025:10.1007/s12035-025-04858-w. [PMID: 40126599 DOI: 10.1007/s12035-025-04858-w] [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: 11/19/2024] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
Acute brain injuries (ABI) caused by various emergencies can lead to structural and functional damage to brain tissue. Common causes include traumatic brain injury, cerebral hemorrhage, ischemic stroke, and heat stroke. Globally, ABI represent a significant portion of neurosurgical cases. Previous studies have emphasized the significant therapeutic potential of stem cell-derived extracellular vesicles (EVs). Recent research indicates that EVs extracted from resident cells in the central nervous system (CNS) also show therapeutic potential following brain injury. Microglia, as innate immune cells of the CNS, respond to changes in the internal environment by altering their phenotype and secreting EVs that impact various CNS cells, including neurons, astrocytes, oligodendrocytes, endothelial cells, neural stem cells (NSCs), and microglia themselves. Notably, under different external stimuli, microglia can either promote neuronal survival, angiogenesis, and myelin regeneration while reducing glial scarring and inflammation, or they can exert opposite effects. This review summarizes and evaluates the current research findings on how microglia-derived EVs influence various CNS cells after ABI under different external stimuli. It analyzes the interaction mechanisms between EVs and resident CNS cells and discusses potential future research directions and clinical applications.
Collapse
Affiliation(s)
- Junjie Gong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Jing Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Jian Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Anqi He
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Bingcheng Ren
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Mingyu Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Kexin Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Yuchi Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Mengyao He
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Yuheng Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China.
| | - Zengguang Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China.
| |
Collapse
|
48
|
Wu J, Li R, Wang J, Zhu H, Ma Y, You C, Shu K. Reactive Astrocytes in Glioma: Emerging Opportunities and Challenges. Int J Mol Sci 2025; 26:2907. [PMID: 40243478 PMCID: PMC11989224 DOI: 10.3390/ijms26072907] [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: 02/14/2025] [Revised: 03/16/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Gliomas are the most prevalent malignant tumors in the adult central nervous system (CNS). Glioblastoma (GBM) accounts for approximately 60-70% of primary gliomas. It is a great challenge to human health because of its high degree of malignancy, rapid progression, short survival time, and susceptibility to recurrence. Owing to the specificity of the CNS, the glioma microenvironment often contains numerous glial cells. Astrocytes are most widely distributed in the human brain and form reactive astrocyte proliferation regions around glioma tissue. In addition, astrocytes are activated under pathological conditions and regulate tumor and microenvironmental cells through cell-to-cell contact or the secretion of active substances. Therefore, astrocytes have attracted attention as important components of the glioma microenvironment. Here, we focus on the mechanisms of reactive astrocyte activation under glioma conditions, their contribution to the mechanisms of glioma genesis and progression, and their potential value as targets for clinical intervention in gliomas.
Collapse
Affiliation(s)
| | | | | | | | | | - Chao You
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095, Jie Fang Avenue, Qiao Kou District, Wuhan 430030, China; (J.W.); (J.W.); (H.Z.); (Y.M.)
| | - Kai Shu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095, Jie Fang Avenue, Qiao Kou District, Wuhan 430030, China; (J.W.); (J.W.); (H.Z.); (Y.M.)
| |
Collapse
|
49
|
Kour D, Khajuria P, Sharma K, Sharma A, Sharma A, Ali SM, Wazir P, Ramajayan P, Sawant SD, Nandi U, Ahmed Z, Kumar A. Isobavachalcone ameliorates Alzheimer disease pathology by autophagy-mediated clearance of amyloid beta and inhibition of NLRP3 inflammasome in primary astrocytes and 5x-FAD mice. Front Pharmacol 2025; 16:1525364. [PMID: 40183098 PMCID: PMC11965660 DOI: 10.3389/fphar.2025.1525364] [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: 11/09/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Background and Aim Alzheimer's disease (AD) progresses with Aβ plaque deposition and neuroinflammation. Given the complexity of AD pathology, single-target therapies have frequently failed in clinical trials. We hypothesized that a multitarget approach could yield better therapeutic outcomes. To this end, we identified isobavachalcone (IBC), a natural compound with dual pharmacological activity in reducing Aβ plaques and neuroinflammation. Experimental Procedure Primary astrocytes were isolated from 3 to 4 days old C57BL/6J mice pups for in-vitro assays, while in-vivo studies were conducted on 5x-FAD mice. Protein alterations were evaluated using ELISA, western blotting, immunocytochemistry, and immunohistochemistry. Behavioral analyses included the radial arm maze, open field, and rotarod tests. Data from all in vitro and in vivo experiments were analyzed by using one-way ANOVA and post-hoc Bonferroni tests. Results In-vitro analyses in astrocytes demonstrated that IBC at 5 and 10 μM concentrations induce AMPK phosphorylation through CAMKK2, promoting autophagy and inhibiting the NLRP3 inflammasome in primary astrocytes. IBC-treated astrocytes exhibited significant clearance of extracellular amyloid beta. Mechanistic studies highlighted autophagy as a key factor in reducing both NLRP3 inflammasome activity and Aβ levels. Two months of treatment of 5x-FAD mice with IBC at 25 and 50 mg/kg significantly improved cognitive functions, as evidenced by enhanced memory and motor performance in behavioral tests. Subsequent brain tissue analysis revealed that IBC upregulated autophagic proteins to reduce the brain's amyloid beta levels, resulting in decreased expression of neuroinflammation markers. Conclusion IBC effectively ameliorates AD pathology through autophagy-mediated clearance of Aβ and suppressing neuroinflammation in 5x-FAD mice.
Collapse
Affiliation(s)
- Dilpreet Kour
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Parul Khajuria
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kuhu Sharma
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Alpa Sharma
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Ankita Sharma
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Syed Mudassir Ali
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Priya Wazir
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - P. Ramajayan
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sanghapal D. Sawant
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Organic Chemsitry Division, CSIR-National Chemical Laboratory, Pune, India
| | - Utpal Nandi
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Zabeer Ahmed
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Ajay Kumar
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
50
|
Thau-Habermann N, Gschwendtberger T, Bodemer C, Petri S. Parthenolide regulates microglial and astrocyte function in primary cultures from ALS mice and has neuroprotective effects on primary motor neurons. PLoS One 2025; 20:e0319866. [PMID: 40100917 PMCID: PMC11918366 DOI: 10.1371/journal.pone.0319866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
Over the last twenty years, the role of microgliosis and astrocytosis in the pathophysiology of neurodegenerative diseases has increasingly been recognized. Dysregulation of microglial and astrocyte properties and function has been described also in the fatal degenerative motor neuron disease amyotrophic lateral sclerosis (ALS). Microglia cells, the immune cells of the nervous system, can either have an immunonegative neurotoxic or immunopositive neuroprotective phenotype. The feverfew plant (Tanacetum parthenium) derived compound parthenolide has been found to be capable of interfering with microglial phenotype and properties. Positive treatment effects were shown in animal models of neurodegenerative diseases like Alzheimer's disease and Parkinson's disease. Now we were able to show that PTL has a modulating effect on primary mouse microglia cells, both wild type and SOD1, causing them to adopt a more neuroprotective potential. Furthermore, we were able to show that PTL, through its positive effect on microglia, also has an indirect positive impact on motor neurons, although PTL itself has no direct effect on these primary motor neurons. The results of our study give reason to consider PTL as a drug candidate for ALS.
Collapse
Affiliation(s)
| | | | - Colin Bodemer
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Susanne Petri
- Department of Neurology, Hannover Medical School, Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
| |
Collapse
|