1
|
Hisaoka-Nakashima K, Tokuda S, Goto T, Yoshii N, Nakamura Y, Ago Y, Morioka N. Hippocampal microglial activation induces cognitive impairment and allodynia through neuronal plasticity changes in male mice with neuropathic pain. Behav Brain Res 2025; 488:115590. [PMID: 40254263 DOI: 10.1016/j.bbr.2025.115590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 04/22/2025]
Abstract
Clinical evidence indicates that cognitive impairment is a common comorbidity of chronic pain, including neuropathic pain, but the mechanism underlying this comorbidity remains unclear. Neuroinflammation plays a critical role in the development of both neuropathic pain and cognitive impairment. A previous study showed that minocycline, an inhibitor of microglia, ameliorated allodynia and cognitive impairment in partial sciatic nerve ligation (PSNL) mice. Therefore, the current study examined a potential role of brain microglia in allodynia and cognitive impairment in male mice with neuropathic pain due to PSNL. Immunohistochemistry of the microglial markers ionized calcium-binding adapter molecule 1 (Iba1), transmembrane protein 119 (TMEM119), and purinergic receptor P2Y12 (P2RY12) was performed to examine microglial status. Two weeks after PSNL, significant microglial activation was observed in the hippocampus and amygdala, but not in the perirhinal cortex. Inhibition of brain-region-specific microglia with a local microinjection of clodronate liposomes was examined to elucidate the involvement of these microglia in PSNL-induced allodynia and cognitive impairment. Local clodronate liposome microinjection to the hippocampus, but not the amygdala, ameliorated allodynia and cognitive impairment. Other changes in the hippocampus of PSNL mice, e.g., decreased hippocampal dendrite length and intersections number, were prevented by microinjection of clodronate liposomes. The current findings suggest hippocampal microglia are related to cognitive impairment and allodynia through neuronal plasticity changes observed in PSNL mice. Blocking hippocampal microglia-mediated neuroinflammation may be a novel approach for reducing comorbidities such as cognitive impairment associated with neuropathic pain.
Collapse
Affiliation(s)
- Kazue Hisaoka-Nakashima
- Department of Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Shintarou Tokuda
- Department of Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Tatsuki Goto
- Department of Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Nanako Yoshii
- Department of Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Yoki Nakamura
- Department of Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Yukio Ago
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Norimitsu Morioka
- Department of Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan.
| |
Collapse
|
2
|
Brioschi S, Han CZ, Colonna M. Drivers and shapers of macrophages specification in the developing brain. Curr Opin Immunol 2025; 94:102558. [PMID: 40239283 DOI: 10.1016/j.coi.2025.102558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025]
Abstract
The brain harbors two major macrophage populations: microglia reside within the brain parenchyma, while border-associated macrophages (BAMs) are situated at central nervous system (CNS) interfaces. BAMs can be further classified into distinct subsets based on their localization: perivascular macrophages surround blood vessels, meningeal macrophages reside in the leptomeninges, dura macrophages in the dura mater, and choroid plexus macrophages are confined to the choroid plexus. The environmental factors and molecular mechanisms driving the specification of these macrophage populations are still being elucidated. Deciphering the communication pathways between CNS macrophages and their tissue niches during development, homeostasis, and pathologic conditions offers significant potential for treating a wide range of brain disorders, from neurodevelopmental and neuroinflammatory diseases to neurovascular and neurodegenerative conditions. With this short review, we will address the current understanding and knowledge gaps in the field, as well as the future directions for the upcoming years.
Collapse
Affiliation(s)
- Simone Brioschi
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO 63110, USA
| | - Claudia Z Han
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine in Saint Louis, Saint Louis, MO 63110, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine in Saint Louis, Saint Louis, MO 63110, USA.
| |
Collapse
|
3
|
Koh Y, Vázquez-Rosa E, Gao F, Li H, Chakraborty S, Tripathi SJ, Barker S, Bud Z, Bangalore A, Kandjoze UP, León-Alvarado RA, Sridharan PS, Cordova BA, Yu Y, Hyung J, Fang H, Singh S, Katabathula R, LaFramboise T, Kasturi L, Lutterbaugh J, Beard L, Cordova E, Cintrón-Pérez CJ, Franke K, Fragoso MF, Miller E, Indrakumar V, Noel KL, Dhar M, Ajroud K, Zamudio C, Lopes FBTP, Bambakidis E, Zhu X, Wilson B, Flanagan ME, Gefen T, Fujioka H, Fink SP, Desai AB, Dawson D, Williams NS, Kim YK, Ready JM, Paul BD, Shin MK, Markowitz SD, Pieper AA. Inhibiting 15-PGDH blocks blood-brain barrier deterioration and protects mice from Alzheimer's disease and traumatic brain injury. Proc Natl Acad Sci U S A 2025; 122:e2417224122. [PMID: 40397680 DOI: 10.1073/pnas.2417224122] [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: 08/24/2024] [Accepted: 04/17/2025] [Indexed: 05/23/2025] Open
Abstract
Alzheimer's disease (AD) and traumatic brain injury (TBI) are currently untreatable neurodegenerative disorders afflicting millions of people worldwide. These conditions are pathologically related, and TBI is one of the greatest risk factors for AD. Although blood-brain barrier (BBB) disruption drives progression of both AD and TBI, strategies to preserve BBB integrity have been hindered by lack of actionable targets. Here, we identify 15-hydroxyprostaglandin dehydrogenase (15-PGDH), an enzyme that catabolizes eicosanoids and other anti-inflammatory mediators, as a therapeutic candidate that protects the BBB. We demonstrate that 15-PGDH is enriched in BBB-associated myeloid cells and becomes markedly elevated in human and mouse models of AD and TBI, as well as aging, another major risk factor for AD. Pathological increase in 15-PGDH correlates with pronounced oxidative stress, neuroinflammation, and neurodegeneration, alongside profound BBB structural degeneration characterized by astrocytic endfeet swelling and functional impairment. Pharmacologic inhibition or genetic reduction of 15-PGDH in AD and TBI models strikingly mitigates oxidative damage, suppresses neuroinflammation, and restores BBB integrity. Most notably, inhibiting 15-PGDH not only halts neurodegeneration but also preserves cognitive function at levels indistinguishable from healthy controls. Remarkably, these neuroprotective effects in AD are achieved without affecting amyloid pathology, underscoring a noncanonical mechanism for treating AD. In a murine microglia cell line exposed to amyloid beta oligomer, major protection was demonstrated by multiple anti-inflammatory substrates that 15-PGDH degrades. Thus, our findings position 15-PGDH inhibition as a broad-spectrum strategy to protect the BBB and thereby preserve brain health and cognition in AD and TBI.
Collapse
Affiliation(s)
- Yeojung Koh
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Department of Pathology, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - Edwin Vázquez-Rosa
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Department of Pathology, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - Farrah Gao
- Department of Genetics and Genome Sciences School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Hongyun Li
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
| | - Suwarna Chakraborty
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21205
| | - Sunil Jamuna Tripathi
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21205
| | - Sarah Barker
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Department of Pathology, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - Zea Bud
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Frances Payne Bolton School of Nursing, Case Western Reserve University, Cleveland, OH 44106
| | - Anusha Bangalore
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Uapingena P Kandjoze
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Department of Psychology, Neuroscience Program Earlham College, Richmond, IN 47374
| | - Rose A León-Alvarado
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Department of Psychology, Neuroscience Program Earlham College, Richmond, IN 47374
| | - Preethy S Sridharan
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Brittany A Cordova
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
| | - Youngmin Yu
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606
| | - Jiwon Hyung
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - Hua Fang
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Hathaway Brown School, Shaker Heights, OH 44122
- Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Salendra Singh
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute Cleveland Clinic, Cleveland, OH 44195
| | | | - Thomas LaFramboise
- Department of Genetics and Genome Sciences School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
| | - Lakshmi Kasturi
- Department of Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - James Lutterbaugh
- Department of Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - Lydia Beard
- Department of Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - Erika Cordova
- Department of Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - Coral J Cintrón-Pérez
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - Kathryn Franke
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | | | - Emiko Miller
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Vidya Indrakumar
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Kamryn L Noel
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Matasha Dhar
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - Kaouther Ajroud
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Department of Pathology, Northwestern University, Chicago, IL 60611
| | - Carlos Zamudio
- Department of Pathology, Northwestern University, Chicago, IL 60611
| | - Filipa Blasco Tavares Pereira Lopes
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Center for Proteomics and Bioinformatics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Evangeline Bambakidis
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Northwestern University Weinberg College of Arts and Sciences, Chicago, IL 60208
| | - Xiongwei Zhu
- Department of Pathology, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - Brigid Wilson
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Department of Infectious Diseases and HIV Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Margaret E Flanagan
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Department of Pathology, Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX 78229
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Hisashi Fujioka
- Cryo-Electron Microscopy Core Case, School of Medicine, Western Reserve University, Cleveland, OH 44016
| | - Stephen P Fink
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
| | - Amar B Desai
- Department of Pathology, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
| | - Dawn Dawson
- Department of Pathology, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Young-Kwang Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Joseph M Ready
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Bindu D Paul
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21205
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21205
- The Solomon H. Snyder Department of Neuroscience, School of Medicine Johns Hopkins University, Baltimore, MD 21205
- Lieber Institute for Brain Development, Baltimore, MD 21205
| | - Min-Kyoo Shin
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Sanford D Markowitz
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
- Department of Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106
| | - Andrew A Pieper
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Department of Pathology, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| |
Collapse
|
4
|
Chen W, Liu X, Muñoz VR, Kahn CR. Loss of insulin signaling in microglia impairs cellular uptake of Aβ and neuroinflammatory response exacerbating AD-like neuropathology. Proc Natl Acad Sci U S A 2025; 122:e2501527122. [PMID: 40388612 DOI: 10.1073/pnas.2501527122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 04/07/2025] [Indexed: 05/21/2025] Open
Abstract
Insulin receptors are present on cells throughout the body, including the brain. Dysregulation of insulin signaling in neurons and astrocytes has been implicated in altered mood, cognition, and the pathogenesis of Alzheimer's disease (AD). To define the role of insulin signaling in microglia, the primary phagocytes in the brain critical for maintenance and damage repair, we created mice with an inducible microglia-specific insulin receptor knockout (MG-IRKO). RiboTag profiling of microglial mRNAs revealed that loss of insulin signaling results in alterations of gene expression in pathways related to innate immunity and cellular metabolism. In vitro, loss of insulin signaling in microglia results in metabolic reprogramming with an increase in glycolysis and impaired uptake of Aβ. In vivo, MG-IRKO mice exhibit alterations in mood and social behavior, and when crossed with the 5xFAD mouse model of AD, the resultant mice exhibit increased levels of Aβ plaque and elevated neuroinflammation. Thus, insulin signaling in microglia plays a key role in microglial cellular metabolism and the ability of the cells to take up Aβ, such that reduced insulin signaling in microglia alters mood and social behavior and accelerates AD pathogenesis. Together, these data indicate key roles of insulin action in microglia and the potential of targeting insulin signaling in microglia in treatment of AD.
Collapse
Affiliation(s)
- Wenqiang Chen
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02215
- Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev 2730, Denmark
| | - Xiangyu Liu
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02215
| | - Vitor Rosetto Muñoz
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02215
- Laboratory of Molecular Biology of Exercise, University of Campinas, Limeira, São Paulo 13484-350, Brazil
| | - C Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02215
| |
Collapse
|
5
|
Chen J, Huang J, Han T, Kojima N. Chronic Stress Modulates Microglial Activation Dynamics, Shaping Priming Responses to Subsequent Stress. Brain Sci 2025; 15:534. [PMID: 40426704 DOI: 10.3390/brainsci15050534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2025] [Revised: 05/19/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
(1) Background: The high recurrence rate and individual differences in stress susceptibility contribute to the diverse symptoms of depression, making full recovery and relapse prevention challenging. Emerging evidence suggests that fluctuations in microglial activity are closely linked to depression progression under chronic stress exposure. Changes in the brain microenvironment can elicit microglial priming, enhancing their sensitivity to external stimuli. However, few studies have longitudinally examined how microglial characteristics evolve throughout depression progression. (2) Methods: In this study, we investigated microglial morphological changes and their responses to acute stress at different stages of depression using the chronic unpredictable mild stress (CUMS) paradigm in mice. (3) Results: Our findings reveal that in the dentate gyrus, microglial activation indices, including cell number and morphology, exhibit distinct dynamic patterns depending on CUMS exposure duration. Notably, after 2 and 4 weeks of CUMS exposure followed by acute stress re-exposure, microglia display opposing response patterns. In contrast, after 6 weeks of CUMS exposure, primed microglia exhibit dysfunction, failing to respond to acute stress. Notably, depressive behaviors are not prominent after 2 weeks of CUMS exposure but become more pronounced after 4 and 6 weeks of exposure. Additionally, regardless of CUMS duration, body weight demonstrates an intrinsic capacity to normalize after stress cessation. (4) Conclusions: These findings suggest that microglial priming responses are state-dependent, either enhancing or suppressing secondary stimulus responses, or exceeding physiological limits, thereby preventing further activation. This study provides novel insights into the role of microglial priming in stress vulnerability and its contribution to depression progression.
Collapse
Affiliation(s)
- Junyu Chen
- Laboratory of Molecular Neurobiology, Faculty of Life Sciences, Toyo University, Saitama 351-8510, Japan
- Super-Network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Jiacheng Huang
- Laboratory of Molecular Neurobiology, Faculty of Life Sciences, Toyo University, Saitama 351-8510, Japan
- Department of Histology and Cell Biology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8640, Japan
| | - Taolei Han
- Laboratory of Molecular Neurobiology, Faculty of Life Sciences, Toyo University, Saitama 351-8510, Japan
| | - Nobuhiko Kojima
- Laboratory of Molecular Neurobiology, Faculty of Life Sciences, Toyo University, Saitama 351-8510, Japan
- Research Center for Biomedical Engineering, Toyo University, Saitama 351-8510, Japan
| |
Collapse
|
6
|
Wang X, Sun Y, Yu H, Xue C, Pei X, Chen Y, Guan Y. The regulation of microglia by aging and autophagy in multiple sclerosis. Pharmacol Res 2025; 216:107786. [PMID: 40398690 DOI: 10.1016/j.phrs.2025.107786] [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: 01/31/2025] [Revised: 04/19/2025] [Accepted: 05/17/2025] [Indexed: 05/23/2025]
Abstract
Multiple sclerosis (MS) is an inflammatory disease that is often characterized by the development of irreversible clinical disability. Age is a strong risk factor that is strongly associated with the clinical course and progression of MS. Several lines of evidence suggest that with aging, microglia have an aging-related gene expression signature and are close to disease-associated microglia (DAM), which exhibit decreased phagocytosis but increased production of inflammatory factors. The gene expression signatures of microglia in MS overlap with those in aging, inflammation and DAM. Moreover, the clearance of damaged myelin by microglia is impaired in the aged brain. Autophagy is a cellular process that decreases in activity with age. In this review, we provide an overview of the role of autophagy and aging in MS. We describe the impact of autophagy and aging on microglial activation in MS and the molecules involved in autophagy and aging, which are related to the phagocytosis and activation of microglia. We propose that a decrease in autophagy in microglia occurs with aging, leading to a decrease in phagocytosis. Decreases in phagocytosis and increases in the production of inflammatory factors by microglia contribute to chronic inflammation in the aged brain and disease progression in MS. Thus, the modulation of autophagy in microglia serves as a potential therapeutic target for MS.
Collapse
Affiliation(s)
- Xiying Wang
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ye Sun
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haojun Yu
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunran Xue
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuzhong Pei
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Chen
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yangtai Guan
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
7
|
Bougnères P, Le Stunff C. Revisiting the Pathogenesis of X-Linked Adrenoleukodystrophy. Genes (Basel) 2025; 16:590. [PMID: 40428412 DOI: 10.3390/genes16050590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2025] [Revised: 05/11/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND X-ALD is a white matter (WM) disease caused by mutations in the ABCD1 gene encoding the transporter of very-long-chain fatty acids (VLCFAs) into peroxisomes. Strikingly, the same ABCD1 mutation causes either devastating brain inflammatory demyelination during childhood or, more often, progressive spinal cord axonopathy starting in middle-aged adults. The accumulation of undegraded VLCFA in glial cell membranes and myelin has long been thought to be the central mechanism of X-ALD. METHODS This review discusses studies in mouse and drosophila models that have modified our views of X-ALD pathogenesis. RESULTS In the Abcd1 knockout (KO) mouse that mimics the spinal cord disease, the late manifestations of axonopathy are rapidly reversed by ABCD1 gene transfer into spinal cord oligodendrocytes (OLs). In a peroxin-5 KO mouse model, the selective impairment of peroxisomal biogenesis in OLs achieves an almost perfect phenocopy of cerebral ALD. A drosophila knockout model revealed that VLCFA accumulation in glial myelinating cells causes the production of a toxic lipid able to poison axons and activate inflammatory cells. Other mouse models showed the critical role of OLs in providing energy substrates to axons. In addition, studies on microglial changing substates have improved our understanding of neuroinflammation. CONCLUSIONS Animal models supporting a primary role of OLs and axonal pathology and a secondary role of microglia allow us to revisit of X-ALD mechanisms. Beyond ABCD1 mutations, pathogenesis depends on unidentified contributors, such as genetic background, cell-specific epigenomics, potential environmental triggers, and stochasticity of crosstalk between multiple cell types among billions of glial cells and neurons.
Collapse
Affiliation(s)
- Pierre Bougnères
- MIRCen Institute, Commissariat à l'Energie Atomique, Laboratoire des Maladies Neurodégénératives, 92260 Fontenay-aux-Roses, France
- NEURATRIS, 92260 Fontenay-aux-Roses, France
- Therapy Design Consulting, 94300 Vincennes, France
| | - Catherine Le Stunff
- MIRCen Institute, Commissariat à l'Energie Atomique, Laboratoire des Maladies Neurodégénératives, 92260 Fontenay-aux-Roses, France
- NEURATRIS, 92260 Fontenay-aux-Roses, France
- UMR1195 Inserm, University Paris Saclay, 94270 Le Kremlin-Bicêtre, France
| |
Collapse
|
8
|
Aisenberg WH, O'Brien CA, Sangster M, Yaqoob F, Zhang Y, Temsamrit B, Thom S, Gosse L, Chaluvadi S, Elfayomi B, Lee G, Polam V, Levitt EM, Liu G, Lombroso SI, Nemec KM, Clowry G, Nieves C, Rawat P, Church E, Martinez D, Shoffler C, Kancheva D, Petucci C, Taylor D, Kofler J, Erskine D, Movahedi K, Bennett ML, Bennett FC. Direct microglia replacement reveals pathologic and therapeutic contributions of brain macrophages to a monogenic neurological disease. Immunity 2025; 58:1254-1268.e9. [PMID: 40311614 PMCID: PMC12078009 DOI: 10.1016/j.immuni.2025.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 12/20/2024] [Accepted: 03/26/2025] [Indexed: 05/03/2025]
Abstract
Krabbe disease, also named globoid cell (GC) leukodystrophy (GLD) for its distinct lipid-laden macrophages, is a severe leukodystrophy caused by galactosylceramidase (GALC) mutations. Hematopoietic stem cell transplant (HSCT) ameliorates disease and is associated with central nervous system (CNS) engraftment of GALC+ donor macrophages. Yet, the role of macrophages in GLD pathophysiology and HSCT remains unclear. Using single-cell sequencing, we revealed early interferon response signatures that preceded progressively severe macrophage dyshomeostasis and identified a molecular signature of GCs, which we validated in human brain specimens. Genetic depletion and direct microglia replacement by CNS monocyte injection rapidly replaced >80% of endogenous microglia with healthy macrophages in the twitcher (GalcW355∗) mouse model of GLD. Perinatal microglia replacement completely normalized transcriptional signatures, rescued histopathology, and doubled average survival. Overall, we uncovered distinct forms of microglial dysfunction and evidence that direct, CNS-limited microglia replacement improves a monogenic neurodegenerative disease, identifying a promising therapeutic target.
Collapse
Affiliation(s)
- William H Aisenberg
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carleigh A O'Brien
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Madison Sangster
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Fazeela Yaqoob
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuanchao Zhang
- Department of Biomedical Informatics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Brian Temsamrit
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Searlait Thom
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Luca Gosse
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Sai Chaluvadi
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bilal Elfayomi
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gavin Lee
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vidhur Polam
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eli M Levitt
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gary Liu
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sonia I Lombroso
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kelsey M Nemec
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gavin Clowry
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Cassaundra Nieves
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Priyanka Rawat
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Emily Church
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Martinez
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Clarissa Shoffler
- Penn Metabolomics Core, Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daliya Kancheva
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Christopher Petucci
- Penn Metabolomics Core, Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Deanne Taylor
- Department of Biomedical Informatics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Julia Kofler
- Division of Neuropathology, Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel Erskine
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Kiavash Movahedi
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mariko L Bennett
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - F Chris Bennett
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
9
|
O'Reilly ML, Wulf MJ, Connors TM, Jin Y, Bearoff F, Bouyer J, Kortagere S, Bethea JR, Tom VJ. Microglial IKKβ Alters Central and Peripheral Immune Activity at Distinct Time Points After Spinal Cord Injury. Glia 2025. [PMID: 40346894 DOI: 10.1002/glia.70030] [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: 08/05/2024] [Revised: 04/19/2025] [Accepted: 04/22/2025] [Indexed: 05/12/2025]
Abstract
After high-level spinal cord injury (SCI), persistently reactive microglia drive widespread plasticity throughout the neuraxis. Plasticity in the thoracolumbar cord, a region corresponding to the spinal sympathetic reflex (SSR) circuit, contributes to the development of sympathetic dysfunction and associated immune disorders. The transcription factor NF-κB is activated after SCI, promoting a pro-inflammatory loop by driving the expression of inflammatory mediators which further activate NF-κB signaling. We hypothesize that microglial NF-κB signaling via IKKβ modulates microglial activity, impacting central and peripheral immune activity related to the SSR circuit post-SCI. We assessed the effect of deleting canonical IKKβ in CNS-resident microglia, its impact on microglial activation, polarization, central transcriptional activity, and peripheral immune activity at 1- and 4-week post-SCI (wpi). Transcriptomic analyses reveal microglial IKKβ influences immune-related pathways in the thoracolumbar cord at 1 wpi. We show that inhibition of microglial NF-κB signaling via deletion of the activator IKKβ mitigates injury-induced increases in "proinflammatory" M1 microglia in the thoracolumbar cord at 4 wpi and increases the quantity of splenocytes at 1 wpi. This study advances our understanding of how microglial IKKβ signaling shapes the neuroimmune response and a peripheral immune organ after SCI.
Collapse
Affiliation(s)
- Micaela L O'Reilly
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Mariah J Wulf
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Theresa M Connors
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Ying Jin
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Frank Bearoff
- Department of Neurology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Julien Bouyer
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Sandhya Kortagere
- Department of Microbiology & Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - John R Bethea
- Department of Anatomy and Cell Biology, George Washington University, Washington, DC, USA
| | - Veronica J Tom
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
10
|
Liu J, Wang Z, Liang W, Zhang Z, Deng Y, Chen X, Hou Z, Xie Y, Wang Q, Li Y, Bai C, Li D, Mo F, Wang H, Wang D, Yuan J, Wang Y, Teng ZQ, Hu B. Microglial TMEM119 binds to amyloid-β to promote its clearance in an Aβ-depositing mouse model of Alzheimer's disease. Immunity 2025:S1074-7613(25)00181-5. [PMID: 40373772 DOI: 10.1016/j.immuni.2025.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 01/19/2025] [Accepted: 04/15/2025] [Indexed: 05/17/2025]
Abstract
The progression of Alzheimer's disease (AD) involves temporal dynamics of microglial activation. Restoring or maintaining microglial homeostasis has emerged as a promising therapeutic strategy to combat AD. Transmembrane protein 119 (TMEM119) is a homeostatic marker of microglia but has not been fully studied under AD pathological conditions. Here, we observed that amyloid-beta (Aβ) induced a decrease in TMEM119 expression in microglia, and TMEM119 deficiency increased AD progression in the 5×FAD mouse model. TMEM119 bound to Aβ oligomers and recruited low-density lipoprotein receptor 1, which in turn degraded TMEM119 itself. Overexpression of TMEM119 in microglia enhanced their phagocytic activity and alleviated cognitive deficits in 5xFAD mice. Administration of the small molecules Kartogenin and SRI-011381, which we found enhanced TMEM119 expression, substantially promoted Aβ clearance and improved cognitive function in AD mice, even during the mid-stage of the disease. These findings identify TMEM119 as a promising therapeutic target for AD.
Collapse
Affiliation(s)
- Jing Liu
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhimeng Wang
- School of Pharmaceutical Sciences, Tsinghua University, Tsinghua-Peking Center for Life Sciences, Beijing 100084, China; Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing 100191, China
| | - Wenwen Liang
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing 100191, China
| | - Zhenhao Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yusen Deng
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaowei Chen
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zongren Hou
- Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuanzhi Xie
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Qi Wang
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing 100191, China
| | - Yuan Li
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing 100191, China
| | - Chaobo Bai
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing 100191, China
| | - Da Li
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Fan Mo
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Huinan Wang
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Dongmei Wang
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Junliang Yuan
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing 100191, China.
| | - Yukai Wang
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China.
| | - Zhao-Qian Teng
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; Medical School, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Baoyang Hu
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; Medical School, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
11
|
Naydovich LR, Orthmann-Murphy JL, Markowitz CE. Beyond relapses: How BTK inhibitors are shaping the future of progressive MS treatment. Neurotherapeutics 2025:e00602. [PMID: 40345950 DOI: 10.1016/j.neurot.2025.e00602] [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: 12/24/2024] [Revised: 04/18/2025] [Accepted: 04/21/2025] [Indexed: 05/11/2025] Open
Abstract
Multiple sclerosis is a biologically and clinically heterogenous inflammatory demyelinating disease, driven by relapsing and progressive mechanisms, all individuals experiencing varying degrees of both. Existing highly effective therapies target peripheral inflammation and reduce relapse rates but have limited efficacy in progressive MS due to poor blood-brain barrier penetration and inability to address neurodegeneration. Bruton's tyrosine kinase (BTK) inhibitors represent an emerging therapeutic class offering a novel mechanism targeting BTK, which is expressed by both B cells and myeloid cells, including microglia within the CNS. Pre-clinical, Phase II, and Phase III clinical trials have demonstrated promising results in modulating progressive disease in both relapsing and non-relapsing MS patients. In contrast, the evidence regarding impact on relapse biology remains mixed and somewhat inconclusive. This review highlights gaps in current therapeutic strategies, examines the latest evidence for the efficacy and safety of BTK inhibitors in MS, and explores the future landscape of MS treatment.
Collapse
Affiliation(s)
- Laura R Naydovich
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.
| | | | - Clyde E Markowitz
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.
| |
Collapse
|
12
|
Nyame K, Xiong J, Alsohybe HN, de Jong APH, Peña IV, de Miguel R, Brummelkamp TR, Hartmann G, Nijman SMB, Raaben M, Simcox JA, Blomen VA, Abu-Remaileh M. PLA2G15 is a BMP hydrolase and its targeting ameliorates lysosomal disease. Nature 2025:10.1038/s41586-025-08942-y. [PMID: 40335701 DOI: 10.1038/s41586-025-08942-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 03/25/2025] [Indexed: 05/09/2025]
Abstract
Lysosomes catabolize lipids and other biological molecules, maintaining cellular and organismal homeostasis. Bis(monoacylglycero)phosphate (BMP), a major lipid constituent of intralysosomal vesicles, stimulates lipid-degrading enzymes and is altered in various human conditions, including neurodegenerative diseases1,2. Although lysosomal BMP synthase was recently discovered3, the enzymes mediating BMP turnover remain elusive. Here we show that lysosomal phospholipase PLA2G15 is a physiological BMP hydrolase. We further demonstrate that the resistance of BMP to lysosomal hydrolysis arises from its unique sn2, sn2' esterification position and stereochemistry, as neither feature alone confers resistance. Purified PLA2G15 catabolizes most BMP species derived from cell and tissue lysosomes. Furthermore, PLA2G15 efficiently hydrolyses synthesized BMP stereoisomers with primary esters, challenging the long-held thought that BMP stereochemistry alone ensures resistance to acid phospholipases. Conversely, BMP with secondary esters and S,S stereoconfiguration is stable in vitro and requires acyl migration for hydrolysis in lysosomes. Consistent with our biochemical data, PLA2G15-deficient cells and tissues accumulate several BMP species, a phenotype reversible by supplementing wild-type PLA2G15 but not its inactive mutant. Targeting PLA2G15 reduces the cholesterol accumulation in fibroblasts of patients with Niemann-Pick disease type C1 and significantly ameliorates disease pathologies in Niemann-Pick disease type C1-deficient mice, leading to an extended lifespan. Our findings established the rules governing BMP stability in lysosomes and identified PLA2G15 as a lysosomal BMP hydrolase and a potential target for therapeutic intervention in neurodegenerative diseases.
Collapse
Affiliation(s)
- Kwamina Nyame
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA, USA
- Department of Biochemistry, Stanford University, Stanford, CA, USA
| | - Jian Xiong
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA, USA
| | - Hisham N Alsohybe
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA, USA
| | | | - Isabelle V Peña
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA, USA
| | | | - Thijn R Brummelkamp
- Scenic Biotech, Science Park 301, Amsterdam, The Netherlands
- Oncode Institute, Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Guido Hartmann
- Scenic Biotech, Science Park 301, Amsterdam, The Netherlands
| | | | - Matthijs Raaben
- Scenic Biotech, Science Park 301, Amsterdam, The Netherlands
| | - Judith A Simcox
- Howard Hughes Medical Institute, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Monther Abu-Remaileh
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA.
- Department of Genetics, Stanford University, Stanford, CA, USA.
- Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA, USA.
- The Phil & Penny Knight Initiative for Brain Resilience at the Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
13
|
Islam R, Choudhary HH, Zhang F, Mehta H, Yoshida J, Thomas A, Hanafy K. Macrophage Lyn Kinase Is a Sex-Specific Regulator of Post-Subarachnoid Hemorrhage Neuroinflammation. J Am Heart Assoc 2025; 14:e039409. [PMID: 40281655 DOI: 10.1161/jaha.124.039409] [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: 10/15/2024] [Accepted: 03/28/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Lyn kinase is a member of the Src family of tyrosine kinases, primarily known for its role in regulating immune cell signaling. It can phosphorylate and modulate the activity of various proteins involved in immune responses, including Toll-like receptor 4 (TLR4). TLR4-mediated inflammatory pathways have been extensively studied; however, the sex-specific interaction of TLR4 and Lyn in neuroinflammation after aneurysmal subarachnoid hemorrhage (SAH) has yet to be investigated. SAH occurs due to a ruptured aneurysm, and the consequences often lead to neuroinflammation and cognitive impairments. In our study, we investigated the sex-specific involvement of Lyn kinase in regulating TLR4 signaling to understand the TLR4-mediated inflammatory response after SAH. METHODS Cell-specific Lyn knockout mice of both sexes were used for this study. Wild-type and conditional knockout mouse brains were analyzed by multicolor flow cytometry, immunohistochemistry, and western blotting at postoperative day 7 following SAH surgery. An unbiased spatial transcriptomic analysis was performed with the frozen mouse brain tissues. A 3-dimensional brain stroke model and cerebrospinal fluid samples of patients with SAH were also used for this study. RESULTS Our overall animal and patient data from flow cytometry, immunohistochemistry, western blot, cognitive function tests, and spatial transcriptomic data revealed that Lyn kinase is a sex-specific regulator in inflammatory cytokine production, red blood cell phagocytosis, neuronal apoptosis, and cognitive function, as well as a negative regulator of TLR4 signaling pathways. CONCLUSIONS Our results highlight sex-specific modulation of Lyn kinase activity in TLR4 signaling after hemorrhagic stroke and indicate that successful treatment of neuroinflammation may require sex-specific treatments.
Collapse
Affiliation(s)
- Rezwanul Islam
- Cooper Medical School of Rowan University Camden NJ USA
- Department of Neurology Cooper University Health Care Camden NJ USA
| | | | - Feng Zhang
- Cooper Medical School of Rowan University Camden NJ USA
- Department of Neurology Cooper University Health Care Camden NJ USA
| | - Hritik Mehta
- Cooper Medical School of Rowan University Camden NJ USA
- Department of Neurology Cooper University Health Care Camden NJ USA
| | - Jun Yoshida
- Cooper Medical School of Rowan University Camden NJ USA
| | - Ajith Thomas
- Cooper Medical School of Rowan University Camden NJ USA
- Department of Neurosurgery Cooper University Health Care Camden NJ USA
| | - Khalid Hanafy
- Cooper Medical School of Rowan University Camden NJ USA
- Department of Neurology Cooper University Health Care Camden NJ USA
- Center for Neuroinflammation Cooper Medical School of Rowan University Camden NJ USA
| |
Collapse
|
14
|
Zhao W, Zhang Z, Xie M, Ding F, Zheng X, Sun S, Du J. Exploring tumor-associated macrophages in glioblastoma: from diversity to therapy. NPJ Precis Oncol 2025; 9:126. [PMID: 40316746 PMCID: PMC12048723 DOI: 10.1038/s41698-025-00920-x] [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: 11/21/2024] [Accepted: 04/22/2025] [Indexed: 05/04/2025] Open
Abstract
Glioblastoma is the most aggressive and lethal cancer of the central nervous system, presenting substantial treatment challenges. The current standard treatment, which includes surgical resection followed by temozolomide and radiation, offers limited success. While immunotherapies, such as immune checkpoint inhibitors, have proven effective in other cancers, they have not demonstrated significant efficacy in GBM. Emerging research highlights the pivotal role of tumor-associated macrophages (TAMs) in supporting tumor growth, fostering treatment resistance, and shaping an immunosuppressive microenvironment. Preclinical studies show promising results for therapies targeting TAMs, suggesting potential in overcoming these barriers. TAMs consist of brain-resident microglia and bone marrow-derived macrophages, both exhibiting diverse phenotypes and functions within the tumor microenvironment. This review delves into the origin, heterogeneity, and functional roles of TAMs in GBM, underscoring their dual roles in tumor promotion and suppression. It also summarizes recent progress in TAM-targeted therapies, which may, in combination with other treatments like immunotherapy, pave the way for more effective and personalized strategies against this aggressive malignancy.
Collapse
Affiliation(s)
- Wenwen Zhao
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhi Zhang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Mingyuan Xie
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Feng Ding
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiangrong Zheng
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shicheng Sun
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jianyang Du
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
15
|
Pan K, Gao Y, Zong H, Zhang Y, Qi Y, Wang H, Chen W, Zhou T, Zhao J, Yin T, Guo H, Wang M, Wang H, Pang T, Zang Y, Li J. Neuronal CCL2 responds to hyperglycaemia and contributes to anxiety disorders in the context of diabetes. Nat Metab 2025; 7:1052-1072. [PMID: 40329008 DOI: 10.1038/s42255-025-01281-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 03/17/2025] [Indexed: 05/08/2025]
Abstract
Anxiety disorders are frequently observed in patients with diabetes and can be associated with several diabetes-related factors. Here we determine that hyperglycaemia is a major cause for the development of anxiety disorders through a C-C motif chemokine ligand 2 (CCL2)-dependent mechanism. By adopting complementary strategies, we demonstrate that neuron-specific (not peripheral) CCL2 mediates anxiety-like behaviours in streptozotocin-induced diabetic mice. Mechanistically, high glucose levels induce Tonicity-responsive enhancer-binding protein (TonEBP)-dependent CCL2 expression in neurons, leading to microglial activation in a paracrine manner. Similar phenotypes are also observed in high-fat diet-induced diabetic mice, independent of insulin signalling. Furthermore, we reveal that neuronal CCL2 in the medial prefrontal cortex and ventral hippocampus synergistically induces anxiety-like behaviours, indicating brain region-specific effects on diabetic mice. Finally, we confirm that the neuronal TonEBP-CCL2 axis and inflammatory pathways are both upregulated in patients with diabetes. Conclusively, neuronal CCL2 is specifically increased by hyperglycaemia and contributes to anxiety disorders, providing additional insights into the link between diabetes and mental health disorders.
Collapse
Affiliation(s)
- Kaijun Pan
- Metabolic Disease Research Center, Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yanan Gao
- Metabolic Disease Research Center, Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Haichao Zong
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yongmei Zhang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yingbei Qi
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Hanlin Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Wengang Chen
- Metabolic Disease Research Center, Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Ting Zhou
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jinwen Zhao
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Tao Yin
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Haoran Guo
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Min Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Hanmin Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Tao Pang
- Metabolic Disease Research Center, Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Yi Zang
- Lingang Laboratory, Shanghai, China.
| | - Jia Li
- Metabolic Disease Research Center, Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China.
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
16
|
Zhang Z, Wang H, Tao B, Shi X, Chen G, Ma H, Peng R, Zhang J. Attenuation of Blood-Brain Barrier Disruption in Traumatic Brain Injury via Inhibition of NKCC1 Cotransporter: Insights into the NF-κB/NLRP3 Signaling Pathway. J Neurotrauma 2025; 42:814-831. [PMID: 39879999 DOI: 10.1089/neu.2023.0580] [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: 01/31/2025] Open
Abstract
Following traumatic brain injury (TBI), inhibition of the Na+-K+-Cl- cotransporter1 (NKCC1) has been observed to alleviate damage to the blood-brain barrier (BBB). However, the underlying mechanism for this effect remains unclear. This study aimed to investigate the mechanisms by which inhibiting the NKCC1 attenuates disruption of BBB integrity in TBI. The TBI model was induced in C57BL/6 mice through a controlled cortical impact device, and an in vitro BBB model was established using Transwell chambers. Western blot (WB) analysis was used to evaluate NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome and nuclear factor-kappaB (NF-κB) pathway proteins. Flow cytometry and transendothelial electrical resistance (TEER) were employed to assess endothelial cell apoptosis levels and BBB integrity. ELISA was utilized to measure cytokines interleukin-1β (IL-1β) and matrix metalloproteinase-9 (MMP-9). Immunofluorescence techniques were used to evaluate protein levels and the nuclear translocation of the rela (p65) subunit. The Evans blue dye leakage assay and the brain wet-dry weight method were utilized to assess BBB integrity and brain swelling. Inhibition of NKCC1 reduced the level of NLRP3 inflammasome and the secretion of IL-1β and MMP-9 in microglia. Additionally, NKCC1 inhibition suppressed the activation of the NF-κB signaling pathway, which in turn decreased the level of NLRP3 inflammasome. The presence of NLRP3 inflammasome in BV2 cells led to compromised BBB integrity within an inflammatory milieu. Following TBI, an upregulation of NLRP3 inflammasome was observed in microglia, astrocytes, vascular endothelial cells, and neurons. Furthermore, inhibiting NKCC1 resulted in a decrease in the positive rate of NLRP3 inflammasome in microglia and the levels of inflammatory cytokines IL-1β and MMP-9 after TBI, which correlated with BBB damage and the development of cerebral edema. These findings demonstrate that the suppression of the NKCC1 cotransporter protein alleviates BBB disruption through the NF-κB/NLRP3 signaling pathway following TBI.
Collapse
Affiliation(s)
- Zehan Zhang
- Department of Neurosurgery, PLA Air Force Hospital of Southern Theatre Command, Guangzhou, China
| | - Hui Wang
- Department of Experimental Pathology, Institute of Radiation Medicine, Beijing, China
| | - Bingyan Tao
- Department of Neurosurgery, 961th Hospital of Joint Logistics Support Force, Qiqihaer, China
| | - Xudong Shi
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Neurosurgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Guilin Chen
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Neurosurgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Hengchao Ma
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of Neurosurgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Ruiyun Peng
- Department of Experimental Pathology, Institute of Radiation Medicine, Beijing, China
| | - Jun Zhang
- Department of Neurosurgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
17
|
Yoblinski AR, Dubey J, Myers T, Sathees N, Volk DW, Fish KN, Seney ML. Brain region and sex differences in human microglia morphology and function. Cereb Cortex 2025; 35:bhaf120. [PMID: 40420495 DOI: 10.1093/cercor/bhaf120] [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: 10/02/2024] [Revised: 02/03/2025] [Accepted: 04/23/2025] [Indexed: 05/28/2025] Open
Abstract
Microglia exhibit complex and dynamic morphology that is linked to function. Altered microglia function has been implicated in multiple diseases of the brain, including elevated phagocytosis of neuronal dendritic spines in schizophrenia. However, understanding the relationship between altered microglia and pathophysiology first requires a clearer picture of microglia morphology in the non-diseased brain, which has yet to be fully established. Here, we deploy immunostaining and confocal microscopy to sample over 1,300 microglia from two prefrontal cortex (PFC) subregions in postmortem human brain (3 males, 3 females). We use Neurolucida 360 to trace the 3-dimensional structure of these microglia and quantify interactions with dendritic spines. We find that PFC microglia in male subjects display overall more complex branching than in female subjects, and subgenual anterior cingulate cortex (sgACC) microglia are more complexly branched with more round somas than those in the dorsolateral PFC (DLPFC), irrespective of sex. Furthermore, a lower proportion of phagocytic burden in sgACC microglia involves engulfment of dendritic spines compared to DLPFC. Overall, our results paint a detailed and nuanced picture of microglia morphology and function in subjects unaffected by psychiatric or neurologic illness that can be used as a benchmark for future studies of the diseased brain.
Collapse
Affiliation(s)
- Andrew R Yoblinski
- Department of Psychiatry and Translational Neuroscience Program, University of Pittsburgh Medical School, 450 Technology Drive Suite 223, Pittsburgh, PA 15213, United States
| | - Jyoti Dubey
- Department of Psychiatry and Translational Neuroscience Program, University of Pittsburgh Medical School, 450 Technology Drive Suite 223, Pittsburgh, PA 15213, United States
| | - Tyler Myers
- Department of Psychiatry and Translational Neuroscience Program, University of Pittsburgh Medical School, 450 Technology Drive Suite 223, Pittsburgh, PA 15213, United States
| | - Nitya Sathees
- Department of Psychiatry and Translational Neuroscience Program, University of Pittsburgh Medical School, 450 Technology Drive Suite 223, Pittsburgh, PA 15213, United States
| | - David W Volk
- Department of Psychiatry and Translational Neuroscience Program, University of Pittsburgh Medical School, 450 Technology Drive Suite 223, Pittsburgh, PA 15213, United States
- VISN 4 Mental Illness Research Education and Clinical Center (MIRECC), VA Pittsburgh Healthcare System, University Drive C, Pittsburgh, PA 15240, United States
| | - Kenneth N Fish
- Department of Psychiatry and Translational Neuroscience Program, University of Pittsburgh Medical School, 450 Technology Drive Suite 223, Pittsburgh, PA 15213, United States
| | - Marianne L Seney
- Department of Psychiatry and Translational Neuroscience Program, University of Pittsburgh Medical School, 450 Technology Drive Suite 223, Pittsburgh, PA 15213, United States
| |
Collapse
|
18
|
Zhuo Y, Xu Y, Qu X, Li Q, Sun M, Gao X, Yuan F, Cao M, Pan B. Research on Peripheral Nerve Aging and Degeneration: Cellular Changes and Mechanism Exploration From the Perspective of Single-Cell Sequencing. Eur J Neurosci 2025; 61:e70129. [PMID: 40317786 DOI: 10.1111/ejn.70129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 05/07/2025]
Abstract
As age increases, there are structural and functional alterations in the peripheral nervous system (PNS), significantly affecting movement, sensation and autonomic function. Understanding the characteristics and mechanisms of PNS aging is crucial for preventing and treating related diseases. This study employed single-cell sequencing technology to analyse the dorsal root ganglia (DRG) and sciatic nerve (SN) of aging rats, in comparison with adult rats. The research investigated the mechanisms underlying PNS aging and degeneration, revealing the transcriptional profiles of various cell types. Significant differences were observed in the proportion of Schwann cells between the DRG and SN of adult and aged rats. The Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Ontology (GO) and Gene Set Enrichment Analysis (GSEA) revealed that pathways related to neurodegeneration were upregulated in Schwann cells. Additionally, lipid metabolism pathways were upregulated in the SN of aged rats, suggesting that certain lipid signalling molecules may influence cell proliferation. Through further re-clustering of myelinating Schwann cells, six distinct subtypes were identified. The anti-aging protein protocadherin 9 (PCDH9) was preliminarily screened and found to be significantly downregulated with age. In vitro experiments confirmed that PCDH9 expression is associated with Schwann cell proliferation and differentiation. By using gene expression analysis and cell type across several age groups, this study offers important insights into the mechanisms of PNS aging and degeneration.
Collapse
Affiliation(s)
- Yuyang Zhuo
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yingcai Xu
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xinzhe Qu
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Qupeng Li
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Maji Sun
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiao Gao
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Feng Yuan
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Menghan Cao
- Center of Clinical Oncology, The Afffliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Bin Pan
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
19
|
Riemann K, von Ahsen J, Böhm T, Schlegel M, Kreuzer M, Fenzl T, Russ H, Parsons CG, Rammes G. GAL-201 as a Promising Amyloid-β-Targeting Small-Molecule Approach for Alzheimer's Disease Treatment: Consistent Effects on Synaptic Plasticity, Behavior and Neuroinflammation. Int J Mol Sci 2025; 26:4167. [PMID: 40362405 PMCID: PMC12071807 DOI: 10.3390/ijms26094167] [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: 03/19/2025] [Revised: 04/17/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Soluble oligomeric forms of Amyloid-β (Aβ) are considered the major toxic species leading to the neurodegeneration underlying Alzheimer's disease (AD). Therefore, drugs that prevent oligomer formation might be promising. The atypical dipeptide GAL-201 is orally bioavailable and interferes as a modulator of Aβ aggregation. It binds to aggregation-prone, misfolded Aβ monomers with high selectivity and affinity, thereby preventing the formation of toxic oligomers. Here, we demonstrate that the previously observed protective effect of GAL-201 on synaptic plasticity occurs irrespective of shortages and post-translational modifications (tested isoforms: Aβ1-42, Aβ(p3-42), Aβ1-40 and 3NTyr(10)-Aβ). Interestingly, the neuroprotective activity of a single dose of GAL-201 was still present after one week and correlated with a prevention of Aβ-induced spine loss. Furthermore, we could observe beneficial effects on spine morphology as well as the significantly reduced activation of proinflammatory microglia and astrocytes in the presence of an Aβ1-42-derived toxicity. In line with these in vitro data, GAL-201 additionally improved hippocampus-dependent spatial learning in the "tgArcSwe" AD mouse model after a single subcutaneous administration. By this means, we observed changes in the deposition pattern: through the clustering of misfolded monomers as off-pathway non-toxic Aβ agglomerates, toxic oligomers are removed. Our results are in line with previously collected preclinical data and warrant the initiation of Investigational New Drug (IND)-enabling studies for GAL-201. By demonstrating the highly efficient detoxification of β-sheet monomers, leading to the neutralization of Aβ oligomer toxicity, GAL-201 represents a promising drug candidate against Aβ-derived pathophysiology present in AD.
Collapse
Affiliation(s)
- Katrin Riemann
- Galimedix Therapeutics Inc., 3704 Calvend Lane, Kensington, MD 20895, USA
- Department of Anesthesiology and Intensive Care, School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Jeldrik von Ahsen
- Department of Anesthesiology and Intensive Care, School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Tamara Böhm
- Department of Anesthesiology and Intensive Care, School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Martin Schlegel
- Department of Anesthesiology and Intensive Care, School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Matthias Kreuzer
- Department of Anesthesiology and Intensive Care, School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Thomas Fenzl
- Department of Anesthesiology and Intensive Care, School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Hermann Russ
- Galimedix Therapeutics Inc., 3704 Calvend Lane, Kensington, MD 20895, USA
| | | | - Gerhard Rammes
- Department of Anesthesiology and Intensive Care, School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| |
Collapse
|
20
|
Han X, Wang D, Chen L, Song H, Zheng X, Zhang X, Zhao S, Liang J, Xu T, Hu Z, Sun L. MSC transplantation ameliorates depression in lupus by suppressing Th1 cell-shaped synaptic stripping. JCI Insight 2025; 10:e181885. [PMID: 40048256 PMCID: PMC12016924 DOI: 10.1172/jci.insight.181885] [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: 04/09/2024] [Accepted: 03/04/2025] [Indexed: 04/23/2025] Open
Abstract
Systemic lupus erythematosus (SLE), an autoimmune disease, can cause psychiatric disorders, particularly depression, via immune activation. Human umbilical cord mesenchymal stromal cell (hUCMSC) transplantation (MSCT) has been shown to ameliorate immune dysfunction in SLE by inducing immune tolerance. However, whether MSCT can relieve the depressive symptoms in SLE remains incompletely understood. Here, we demonstrate that MSCT relieved early-onset depression-like behavior in both genetically lupus-prone (MRL/lpr) and pristane-induced lupus mice by rescuing impaired hippocampal synaptic connectivity. Transplanted hUCMSCs targeted Th1 cell-derived IFN-γ to inhibit neuronal JAK/STAT1 signaling and downstream CCL8 expression, reducing phagocytic microglia apposition to alleviate synaptic engulfment and neurological dysfunction in young (8-week-old) lupus mice. Systemic delivery of exogenous IFN-γ blunted MSCT-mediated alleviation of synaptic loss and depressive behavior in lupus mice, suggesting that the IFN-γ/CCL8 axis may be an effective therapeutic target and that MSCT is a potential therapy for lupus-related depression. In summary, transplanted hUCMSCs can target systemic immunity to ameliorate psychiatric disorders by rescuing synaptic loss, highlighting the active role of neurons as intermediaries between systemic immunity and microglia in this process.
Collapse
Affiliation(s)
- Xiaojuan Han
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Dandan Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Liang Chen
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hua Song
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xiulan Zheng
- School of Pharmacy, Macau University of Science and Technology, Macau, China
| | - Xin Zhang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Shengnan Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jun Liang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Tianshu Xu
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhibin Hu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
21
|
Lv K, Luo Y, Liu T, Xia M, Gong H, Zhang D, Chen X, Jiang X, Liu Y, Liu J, Cai Y, Antonson P, Warner M, Xu H, Gustafsson JÅ, Fan X. Inactivation of microglial LXRβ in early postnatal mice impairs microglia homeostasis and causes long-lasting cognitive dysfunction. Proc Natl Acad Sci U S A 2025; 122:e2410698122. [PMID: 40208947 PMCID: PMC12012545 DOI: 10.1073/pnas.2410698122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 02/28/2025] [Indexed: 04/12/2025] Open
Abstract
Microglia, the largest population of brain immune cells, play an essential role in regulating neuroinflammation by removing foreign materials and debris and in cognition by pruning synapses. Since liver X receptor β (LXRβ) has been identified as a regulator of microglial homeostasis, this study examined whether its removal from microglia affects neuroinflammation and cognitive function. We used a cell-specific tamoxifen-inducible Cre-loxP-mediated recombination to remove LXRβ from microglia specifically. We now report that ablation of LXRβ in microglia in early postnatal life led to a reduction in microglial numbers, distinct morphological changes indicative of microglial activation, and enhanced synapse engulfment accompanied by cognitive deficits. Removal of LXRβ from microglia in adult mice caused no cognitive defects. RNAseq analysis of microglia revealed that loss of LXRβ led to reduced expression of SAll1, a master regulator of microglial homeostasis, while increasing expression of genes associated with microglial activation and CNS disease. This study demonstrates distinctly different functions of microglial LXRβ in developing and adult mice and points to long-term consequences of defective LXRβ signaling in microglia in early life.
Collapse
Affiliation(s)
- Keyi Lv
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing400038, China
| | - Yi Luo
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing400038, China
| | - Tianyao Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing400038, China
| | - Meiling Xia
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing400038, China
| | - Hong Gong
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing400038, China
| | - Dandan Zhang
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing400038, China
| | - Xuan Chen
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing400038, China
| | - Xin Jiang
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing400038, China
| | - Yulong Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing400038, China
| | - Jiayin Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing400038, China
| | - Yulong Cai
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing400038, China
| | - Per Antonson
- Center for Innovative Medicine, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm14186, Sweden
| | - Margaret Warner
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX77204
| | - Haiwei Xu
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing400038, China
| | - Jan-Åke Gustafsson
- Center for Innovative Medicine, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm14186, Sweden
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX77204
| | - Xiaotang Fan
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing400038, China
| |
Collapse
|
22
|
Le LHD, Feidler AM, Rodriguez LC, Cealie M, Plunk E, Li H, Kara-Pabani K, Lamantia C, O'Banion MK, Majewska AK. Noradrenergic signaling controls Alzheimer's disease pathology via activation of microglial β2 adrenergic receptors. Brain Behav Immun 2025; 128:307-322. [PMID: 40245958 DOI: 10.1016/j.bbi.2025.04.022] [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: 04/09/2024] [Revised: 04/07/2025] [Accepted: 04/12/2025] [Indexed: 04/19/2025] Open
Abstract
Norepinephrine (NE) is a potent anti-inflammatory agent in the brain. In Alzheimer's disease (AD), the loss of NE signaling heightens neuroinflammation and exacerbates amyloid pathology. NE inhibits surveillance activity of microglia, the brain's resident immune cells, via their β2 adrenergic receptors (β2ARs). Here, we investigate the role of microglial β2AR signaling in AD pathology in the 5xFAD mouse model of AD. We found that loss of cortical NE projections preceded the degeneration of NE-producing neurons and that microglia in 5xFAD mice, especially those microglia that were associated with plaques, significantly downregulated β2AR expression early in amyloid pathology. Importantly, dampening microglial β2AR signaling worsened plaque load and the associated neuritic damage, while stimulating microglial β2AR signaling attenuated amyloid pathology. Our results suggest that microglial β2AR could be explored as a potential therapeutic target to modify AD pathology.
Collapse
Affiliation(s)
- L H D Le
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY, USA
| | - A M Feidler
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY, USA
| | - L Calcines Rodriguez
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY, USA
| | - M Cealie
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY, USA
| | - E Plunk
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY, USA; Department of Environmental Medicine, University of Rochester, Rochester, NY, USA
| | - H Li
- Medical Scientist Training Program, University of Rochester, Rochester NY, USA
| | - K Kara-Pabani
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY, USA
| | - C Lamantia
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY, USA
| | - M K O'Banion
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY, USA
| | - A K Majewska
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY, USA; Center for Visual Science, University of Rochester, Rochester NY, USA.
| |
Collapse
|
23
|
Shi X, Zhang J, Zhao H, Li H, Zhu J, Xiong H. Differential tissue and cellular distribution of chemokine C-C motif ligand 2 in grey/white matters of healthy and simian immunodeficiency virus infected monkey. Brain Res Bull 2025; 223:111291. [PMID: 40054539 DOI: 10.1016/j.brainresbull.2025.111291] [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: 11/25/2024] [Revised: 02/18/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025]
Abstract
Previous studies have shown that CCL2 concentration is higher in cerebrospinal fluid than in plasma of health and human immunodeficiency virus (HIV) infected individuals, suggesting an extra source of CCL2 in brain. Brain cellular CCL2 has been broadly studied in cultured cells and its in-vivo cellular distribution has been investigated in rodent experimental autoimmune encephalomyelitis model. However, its cellular distribution in grey and white matter (GM, WM) remains elusive. We explored this issue using healthy and simian immunodeficiency virus (SIV) infected monkeys and found: 1) Neurons were a major source of CCL2-like immunoreactivity (CCL2-ir) in normal GM, and corpus callosum (CC) ependyma showed high density of CCL2-ir. 2) Upon SIV infection, CCL2-ir was strikingly raised in GM neurons, and in CC ependyma. 3) Brain vascular-perivascular cells were a large source of CCL2-ir in normal GM and WM, which was relatively larger in CC WM than in GM. 4) Vascular-perivascular CCL2-ir proportional areas were significantly enhanced by SIV infection in both GM and CC WM. 5) Microglia seemed not to express CCL2 in healthy brain. Microglia-marker and CCL2-ir co-labeled cells were significantly increased by SIV infection. 6) A vast of macrophage-like cells were situated along infected CC ependyma, suggesting a large number of monocytes be crossing ependyma, which may be related to establishment of viral reservoir. In conclusion, our study provides valuable insights into the cellular sources and alterations of CCL2 in the monkey brain under normal and SIV-infected conditions, which may promote better understanding of CCL2 in related neurological processes.
Collapse
Affiliation(s)
- Xue Shi
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jingdong Zhang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Huangying Zhao
- Division of Pharmaceutical Science, University of Cincinnati College of Pharmacy, Cincinnati, OH 45267, USA
| | - Hongjun Li
- Department of Radiology, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Junyi Zhu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Molecular Biological Targeted Therapies of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Huangui Xiong
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
24
|
Pickles SR, Gonzalez Bejarano J, Narayan A, Daughrity L, Maroto Cidfuentes C, Reeves MM, Yue M, Castellanos Otero P, Estades Ayuso V, Dunmore J, Song Y, Tong J, DeTure M, Rawlinson B, Castanedes‐Casey M, Dulski J, Cerquera‐Cleves C, Zhang Y, Josephs KA, Dickson DW, Petrucelli L, Wszolek ZK, Prudencio M. TDP-43 Cryptic RNAs in Perry Syndrome: Differences across Brain Regions and TDP-43 Proteinopathies. Mov Disord 2025; 40:662-671. [PMID: 39788898 PMCID: PMC12006891 DOI: 10.1002/mds.30104] [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: 10/10/2024] [Revised: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Perry syndrome (PS) is a rare and fatal hereditary autosomal dominant neurodegenerative disorder caused by mutations in dynactin (DCTN1). PS brains accumulate inclusions positive for ubiquitin, transactive-response DNA-binding protein of 43 kDa (TDP-43), and to a lesser extent dynactin. OBJECTIVES Little is known regarding the contributions of TDP-43, an RNA binding protein that represses cryptic exon inclusion, in PS. Therefore, we sought to identify the degree of TDP-43 dysfunction in two regions of PS brains. METHODS We evaluated the levels of insoluble pTDP-43 and TDP-43-regulated cryptic RNAs and protein in the caudate nucleus and substantia nigra of 7 PS cases, 12 cases of frontotemporal lobar degeneration (FTLD) with TDP-43 pathology, and 11 cognitively healthy controls without TDP-43 pathology. RESULTS Insoluble pTDP-43 protein levels were detected in PS brains to a similar extent in the caudate nucleus and substantia nigra but lower than those in FTLD brains. The caudate nucleus of PS showed accumulation of eight TDP-43-regulated cryptic RNAs (ACTL6B, CAMK2B, STMN2, UNC13A, KCNQ2, ATG4B, GPSM2, and HDGFL2) and cryptic protein (HDGFL2) characteristic of FTLD. Conversely, only one cryptic target, UNC13A, reached significance in the substantia nigra despite similar pTDP-43 levels. CONCLUSION We detected TDP-43 cryptic RNAs and protein in PS caudate nucleus. Given the importance of cryptic exon biology in the development of biomarkers, and the identification of novel targets for therapeutic intervention, it is imperative we understand the consequences of TDP-43 dysfunction across different brain regions and determine the targets that are specific and common to TDP-43 proteinopathies. © 2025 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Sarah R. Pickles
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neurobiology of Disease Graduate Program, Mayo Graduate SchoolMayo Clinic College of MedicineJacksonvilleFloridaUSA
| | | | - Anand Narayan
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | | | | | - Mei Yue
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | - Virginia Estades Ayuso
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neurobiology of Disease Graduate Program, Mayo Graduate SchoolMayo Clinic College of MedicineJacksonvilleFloridaUSA
| | - Judy Dunmore
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Yuping Song
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Jimei Tong
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Michael DeTure
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | | | - Jaroslaw Dulski
- Department of NeurologyMayo ClinicJacksonvilleFloridaUSA
- Division of Neurological and Psychiatric Nursing, Faculty of Health SciencesMedical University of GdanskGdanskPoland
- Neurology DepartmentSt Adalbert Hospital, Copernicus PL Ltd.GdanskPoland
| | - Catalina Cerquera‐Cleves
- Department of Neurosciences, Neurology UnitHospital Universitario San IgnacioBogotaColombia
- CHU de Québec Research Center, Axe NeurosciencesUniversité LavalQuebec CityQuebecCanada
| | - Yongjie Zhang
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neurobiology of Disease Graduate Program, Mayo Graduate SchoolMayo Clinic College of MedicineJacksonvilleFloridaUSA
| | | | - Dennis W. Dickson
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neurobiology of Disease Graduate Program, Mayo Graduate SchoolMayo Clinic College of MedicineJacksonvilleFloridaUSA
| | - Leonard Petrucelli
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neurobiology of Disease Graduate Program, Mayo Graduate SchoolMayo Clinic College of MedicineJacksonvilleFloridaUSA
| | | | - Mercedes Prudencio
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neurobiology of Disease Graduate Program, Mayo Graduate SchoolMayo Clinic College of MedicineJacksonvilleFloridaUSA
| |
Collapse
|
25
|
Weng W, Lin B, Zheng J, Sun Y, Li Z, Chen X, Wang Y, Pan X. Novel application of cycloastragenol target microglia for the treatment of Alzheimer's disease: Evidence from single-cell analysis, network pharmacology and experimental assessment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156502. [PMID: 39970860 DOI: 10.1016/j.phymed.2025.156502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 01/24/2025] [Accepted: 02/11/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Cycloastragenol (CAG), a compound extracted from Astragalus, is known for its telomerase activation and anti-inflammatory, antioxidant properties. However, its potential pharmacological effects on Alzheimer's disease (AD) remain unclear. PURPOSE This study aimed to explore potential targets and molecular mechanisms for the role of CAG in alzheimer's disease (AD) treatment. METHODS CAG was administered to 5 × FAD mice. The senescent cell count was verified by senescence-associated β-galactosidase (SA-β-gal) staining. The impact of CAG on microglial phagocytosis was assessed by in vitro and in vivo assays. The potential targets of CAG were identified by network pharmacology and single-nucleus RNA sequencing (snRNA-seq). The underlying mechanism was validated by molecular docking, surface plasmon resonance (SPR) and western blotting. RESULTS CAG effectively ameliorated cognitive impairments and microglial senescence in 5 × FAD mice. In vivo and in vitro experiments revealed that CAG modulated microglial phagocytic activity and reduced hippocampal Aβ deposition The analysis of single-nucleus RNA sequencing data of AD patients reported 13 microglial targets for AD intervention. Phosphodiesterase 4B (PDE4B) was identified as the target through which CAG regulated microglial activity by utilizing network pharmacology, molecular docking and SPR. Western blotting revealed that the PDE4B/CREB/BDNF pathway may mediate the regulatory effect of CAG. CONCLUSION CAG can enhance microglial phagocytosis and alleviate memory dysfunction and amyloid plaque pathology. Our findings suggest that CAG may regulate microglial function through its interaction with PDE4B, providing a novel therapeutic strategy for AD.
Collapse
Affiliation(s)
- Weipin Weng
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, Fuzhou, China; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China; Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Baoping Lin
- Department of Neurology, The First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Jiahao Zheng
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, Fuzhou, China; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China; Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Yixin Sun
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, Fuzhou, China; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China; Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Zijing Li
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, China
| | - Xiaochun Chen
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, Fuzhou, China; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China; Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, China.
| | - Yanping Wang
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Xiaodong Pan
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, Fuzhou, China; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China; Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
26
|
Stoessel MB, Stowell RD, Lowery RL, Le LHD, Vu AN, Whitelaw BS, Majewska AK. The effects of P2Y12 loss on microglial gene expression, dynamics, and injury response in the cerebellum and cerebral cortex. Brain Behav Immun 2025; 128:99-120. [PMID: 40174868 DOI: 10.1016/j.bbi.2025.03.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/26/2025] [Accepted: 03/29/2025] [Indexed: 04/04/2025] Open
Affiliation(s)
- Mark B Stoessel
- Department of Neuroscience, Delmonte Institute for Neuroscience, University of Rochester, Rochester NY 14642, USA; Neuroscience Graduate Program, University of Rochester, Rochester, NY 14642, USA.
| | - Rianne D Stowell
- Department of Neuroscience, Delmonte Institute for Neuroscience, University of Rochester, Rochester NY 14642, USA.
| | - Rebecca L Lowery
- Department of Neuroscience, Delmonte Institute for Neuroscience, University of Rochester, Rochester NY 14642, USA
| | - Linh H D Le
- Department of Neuroscience, Delmonte Institute for Neuroscience, University of Rochester, Rochester NY 14642, USA; Neuroscience Graduate Program, University of Rochester, Rochester, NY 14642, USA.
| | - Andy N Vu
- Department of Neuroscience, Delmonte Institute for Neuroscience, University of Rochester, Rochester NY 14642, USA
| | - Brendan S Whitelaw
- Department of Neuroscience, Delmonte Institute for Neuroscience, University of Rochester, Rochester NY 14642, USA; Neuroscience Graduate Program, University of Rochester, Rochester, NY 14642, USA; Medical Scientist Training Program, University of Rochester 14642, USA
| | - Ania K Majewska
- Department of Neuroscience, Delmonte Institute for Neuroscience, University of Rochester, Rochester NY 14642, USA; Center for Visual Science, University of Rochester, Rochester, NY 14642, USA.
| |
Collapse
|
27
|
Chen L, Wang W. Microglia-derived sEV: Friend or foe in the pathogenesis of cognitive impairment. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111287. [PMID: 39954801 DOI: 10.1016/j.pnpbp.2025.111287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 01/26/2025] [Accepted: 02/06/2025] [Indexed: 02/17/2025]
Abstract
As immune cells, microglia serve a dual role in cognition. Microglia-derived sEV actively contribute to the development of cognitive impairment by selectively targeting specific cells through various substances such as proteins, RNA, DNA, lipids, and metabolic waste. In recent years, there has been an increasing focus on understanding the pathogenesis and therapeutic potential of sEV. This comprehensive review summarizes the detrimental effects of M1 microglial sEV on pathogenic protein transport, neuroinflammation, disruption of the blood-brain barrier (BBB), neuronal death and synaptic dysfunction in relation to cognitive damage. Additionally, it highlights the beneficial effects of M2 microglia on alleviating cognitive impairment based on evidence from cellular experiments and animal studies. Furthermore, since microglial-secreted sEV can be found in cerebrospinal fluid or cross the BBB into plasma circulation, they play a crucial role in diagnosing cognitive impairment. However, using sEV as biomarkers is still at an experimental stage and requires further clinical validation. Future research should aim to explore the mechanisms underlying microglial involvement in various nervous system disorders to identify novel targets for clinical interventions.
Collapse
Affiliation(s)
- Lilin Chen
- Pulmonary and Critical Care Medicine, Heping District, Shenyang City, Liaoning Province, China
| | - Wei Wang
- Pulmonary and Critical Care Medicine, Heping District, Shenyang City, Liaoning Province, China.
| |
Collapse
|
28
|
Liu N, Jiang Y, Xiu Y, Tortelote GG, Xia W, Wang Y, Li Y, Shi S, Han J, Vidoudez C, Niamnud A, Kilgore MD, Zhou D, Shi M, Graziose SA, Fan J, Katakam PVG, Dumont AS, Wang X. Itaconate restrains acute proinflammatory activation of microglia after traumatic brain injury in mice. Sci Transl Med 2025; 17:eadn2635. [PMID: 40073156 DOI: 10.1126/scitranslmed.adn2635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 08/18/2024] [Accepted: 02/13/2025] [Indexed: 03/14/2025]
Abstract
Traumatic brain injury (TBI) rapidly triggers proinflammatory activation of microglia, contributing to secondary brain damage post-TBI. Although the governing role of energy metabolism in shaping the inflammatory phenotype and function of immune cells has been increasingly recognized, the specific alterations in microglial bioenergetics post-TBI remain poorly understood. Itaconate, a metabolite produced by the enzyme aconitate decarboxylase 1 [IRG1; encoded by immune responsive gene 1 (Irg1)], is a pivotal metabolic regulator in immune cells, particularly in macrophages. Because microglia are macrophages of the brain parenchyma, the IRG1/itaconate pathway likely modulates microglial inflammatory responses. In this study, we explored the role of the IRG1/itaconate pathway in regulating microglial bioenergetics and inflammatory activation post-TBI using a mouse controlled cortical impact (CCI) model. We isolated microglia before and 4 and 12 hours after TBI and observed a swift but transient increase in glycolysis coupled with a prolonged disruption of mitochondrial metabolism after injury. Despite an up-regulation of Irg1 expression, itaconate in microglia declined after TBI. Microglia-specific Irg1 gene knockout (Irg1-Mi-KO) exacerbated metabolic changes, intensified proinflammatory activation and neurodegeneration, and worsened certain long-term neurological deficits. Supplementation with 4-octyl itaconate (OI) reinstated the use and oxidative metabolism of glucose, glutamine, and fatty acid, thereby enhancing microglial bioenergetics post-TBI. OI supplementation also attenuated proinflammatory activation and neurodegeneration and improved long-term neurological outcomes. These results suggest that therapeutically targeting the itaconate pathway could improve microglial energy metabolism and neurological outcomes after TBI.
Collapse
Affiliation(s)
- Ning Liu
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Tulane University Translational Sciences Institute, New Orleans, LA 70112, USA
- Neuroscience Program, Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA
| | - Yinghua Jiang
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Yuwen Xiu
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Giovane G Tortelote
- Department of Pediatrics and Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Winna Xia
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Yingjie Wang
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Yadan Li
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Samuel Shi
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jinrui Han
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Charles Vidoudez
- Harvard Center for Mass Spectrometry, Harvard University, Cambridge, MA 02138, USA
| | - Aim Niamnud
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Mitchell D Kilgore
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Di Zhou
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Mengxuan Shi
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Stephen A Graziose
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jia Fan
- Center for Cellular and Molecular Diagnostics, Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Prasad V G Katakam
- Neuroscience Program, Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Aaron S Dumont
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Xiaoying Wang
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Neuroscience Program, Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
29
|
Rodriguez-Baena FJ, Marquez-Galera A, Ballesteros-Martinez P, Castillo A, Diaz E, Moreno-Bueno G, Lopez-Atalaya JP, Sanchez-Laorden B. Microglial reprogramming enhances antitumor immunity and immunotherapy response in melanoma brain metastases. Cancer Cell 2025; 43:413-427.e9. [PMID: 39919736 DOI: 10.1016/j.ccell.2025.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 11/04/2024] [Accepted: 01/13/2025] [Indexed: 02/09/2025]
Abstract
Melanoma is one of the tumor types with the highest risk of brain metastasis. However, the biology of melanoma brain metastasis and the role of the brain immune microenvironment in treatment responses are not yet fully understood. Using preclinical models and single-cell transcriptomics, we have identified a mechanism that enhances antitumor immunity in melanoma brain metastasis. We show that activation of the Rela/Nuclear Factor κB (NF-κB) pathway in microglia promotes melanoma brain metastasis. Targeting this pathway elicits microglia reprogramming toward a proinflammatory phenotype, which enhances antitumor immunity and reduces brain metastatic burden. Furthermore, we found that proinflammatory microglial markers in melanoma brain metastasis are associated with improved responses to immune checkpoint inhibitors in patients and targeting Rela/NF-κB pathway in mice improves responses to these therapies in the brain, suggesting a strategy to enhance antitumor immunity and responses to immune checkpoint inhibitors in patients with melanoma brain metastasis.
Collapse
Affiliation(s)
| | | | | | - Alba Castillo
- Instituto de Neurociencias (CSIC-UMH), San Juan de Alicante, Spain
| | - Eva Diaz
- MD Anderson Cancer Center International Foundation, Madrid, Spain
| | - Gema Moreno-Bueno
- MD Anderson Cancer Center International Foundation, Madrid, Spain; Instituto de Investigaciones Biomédicas "Sols-Morreale" CSIC-UAM, Madrid, Spain; CIBERONC Centro de Investigación Biomédica en Red de Cancer, ISCIII, Madrid, Spain; Translational Cancer Research Group, Area 3 Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | | | | |
Collapse
|
30
|
Garcia-Gomara M, Legarra-Marcos N, Serena M, Rojas-de-Miguel E, Espelosin M, Marcilla I, Perez-Mediavilla A, Luquin MR, Lanciego JL, Burrell MA, Cuadrado-Tejedor M, Garcia-Osta A. FKBP51 inhibition ameliorates neurodegeneration and motor dysfunction in the neuromelanin-SNCA mouse model of Parkinson's disease. Mol Ther 2025; 33:895-916. [PMID: 39905728 PMCID: PMC11897814 DOI: 10.1016/j.ymthe.2025.01.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 11/16/2024] [Accepted: 01/30/2025] [Indexed: 02/06/2025] Open
Abstract
Parkinson's disease (PD) is characterized by the loss of neuromelanin (NM)-containing dopaminergic (DA) neurons in the substantia nigra (SN) pars compacta (SNpc) and the buildup of α-synuclein (α-syn) inclusions, called Lewy bodies. To investigate the roles of NM and α-syn in DA neuron degeneration, we modeled PD by inducing NM accumulation in a humanized α-syn mouse model (Snca-; PAC-Tg(SNCAWT)) via the expression of human tyrosinase in the SN. We found that this mouse strain develops naturally progressive motor dysfunction and dopaminergic neuronal loss in the SN with aging. Upon tyrosinase injection, NM-containing neurons developed p62 and ubiquitin inclusions. Furthermore, the upregulation of genes associated with microglial activation in the midbrain indicated a role of pro-inflammatory factors in neurodegeneration. Midbrain RNA sequencing confirmed the microglial response and identified Fkbp5 as one of the more dysregulated genes. Next, we showed that FKBP51(51 kDa) was significantly upregulated with aging and in PD human brains. Pharmacological treatment with SAFit2, a potent FKBP51 inhibitor, led to a reduction in ubiquitin-positive inclusions, prevention of neurodegeneration in the SNpc, and improved motor function in NM-SNCAWT mice. These results highlight the critical role of FKBP51 in PD and propose SAFit2 as a promising therapeutic candidate for reducing neurodegeneration in PD.
Collapse
Affiliation(s)
- Marta Garcia-Gomara
- Gene Therapy for CNS Disorders Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, Pamplona, 31008 Navarra, Spain; IdiSNA (Navarra Institute for Health Research), Pamplona, 31008 Navarra, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Irunlarrea 1, 31008 Pamplona, Navarra, Spain
| | - Naroa Legarra-Marcos
- Computational Biology Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, Pamplona, 31008 Navarra, Spain
| | - Maria Serena
- Gene Therapy for CNS Disorders Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, Pamplona, 31008 Navarra, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Irunlarrea 1, 31008 Pamplona, Navarra, Spain
| | - Elvira Rojas-de-Miguel
- IdiSNA (Navarra Institute for Health Research), Pamplona, 31008 Navarra, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Irunlarrea 1, 31008 Pamplona, Navarra, Spain
| | - Maria Espelosin
- Gene Therapy for CNS Disorders Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, Pamplona, 31008 Navarra, Spain; IdiSNA (Navarra Institute for Health Research), Pamplona, 31008 Navarra, Spain
| | - Irene Marcilla
- Gene Therapy for CNS Disorders Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, Pamplona, 31008 Navarra, Spain; IdiSNA (Navarra Institute for Health Research), Pamplona, 31008 Navarra, Spain
| | - Alberto Perez-Mediavilla
- Gene Therapy for CNS Disorders Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, Pamplona, 31008 Navarra, Spain; IdiSNA (Navarra Institute for Health Research), Pamplona, 31008 Navarra, Spain; Biochemistry and Genetics Department, School of Sciences, University of Navarra, Irunlarrea 1, 31008 Pamplona, Navarra, Spain
| | - Maria Rosario Luquin
- IdiSNA (Navarra Institute for Health Research), Pamplona, 31008 Navarra, Spain; Department of Neurology, Clínica Universidad de Navarra, University of Navarra, Avenida Pio XII 36, Pamplona, 31008 Navarra, Spain
| | - Jose Luis Lanciego
- Gene Therapy for CNS Disorders Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, Pamplona, 31008 Navarra, Spain
| | - Maria Angeles Burrell
- IdiSNA (Navarra Institute for Health Research), Pamplona, 31008 Navarra, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Irunlarrea 1, 31008 Pamplona, Navarra, Spain
| | - Mar Cuadrado-Tejedor
- Gene Therapy for CNS Disorders Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, Pamplona, 31008 Navarra, Spain; IdiSNA (Navarra Institute for Health Research), Pamplona, 31008 Navarra, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Irunlarrea 1, 31008 Pamplona, Navarra, Spain.
| | - Ana Garcia-Osta
- Gene Therapy for CNS Disorders Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, Pamplona, 31008 Navarra, Spain; IdiSNA (Navarra Institute for Health Research), Pamplona, 31008 Navarra, Spain.
| |
Collapse
|
31
|
Zhang G, Yao Q, Long C, Yi P, Song J, Wu L, Wan W, Rao X, Lin Y, Wei G, Ying J, Hua F. Infiltration by monocytes of the central nervous system and its role in multiple sclerosis: reflections on therapeutic strategies. Neural Regen Res 2025; 20:779-793. [PMID: 38886942 PMCID: PMC11433895 DOI: 10.4103/nrr.nrr-d-23-01508] [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: 09/06/2023] [Revised: 12/12/2023] [Accepted: 02/18/2024] [Indexed: 06/20/2024] Open
Abstract
Mononuclear macrophage infiltration in the central nervous system is a prominent feature of neuroinflammation. Recent studies on the pathogenesis and progression of multiple sclerosis have highlighted the multiple roles of mononuclear macrophages in the neuroinflammatory process. Monocytes play a significant role in neuroinflammation, and managing neuroinflammation by manipulating peripheral monocytes stands out as an effective strategy for the treatment of multiple sclerosis, leading to improved patient outcomes. This review outlines the steps involved in the entry of myeloid monocytes into the central nervous system that are targets for effective intervention: the activation of bone marrow hematopoiesis, migration of monocytes in the blood, and penetration of the blood-brain barrier by monocytes. Finally, we summarize the different monocyte subpopulations and their effects on the central nervous system based on phenotypic differences. As activated microglia resemble monocyte-derived macrophages, it is important to accurately identify the role of monocyte-derived macrophages in disease. Depending on the roles played by monocyte-derived macrophages at different stages of the disease, several of these processes can be interrupted to limit neuroinflammation and improve patient prognosis. Here, we discuss possible strategies to target monocytes in neurological diseases, focusing on three key aspects of monocyte infiltration into the central nervous system, to provide new ideas for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Guangyong Zhang
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Qing Yao
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Chubing Long
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Pengcheng Yi
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Jiali Song
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Luojia Wu
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Wei Wan
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Xiuqin Rao
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Yue Lin
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Gen Wei
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Jun Ying
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Fuzhou Hua
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
32
|
Zhou L, Wu Z, Yi X, Xie D, Wang J, Wu W. Serum starvation induces cytosolic DNA trafficking via exosome and autophagy-lysosome pathway in microglia. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119905. [PMID: 39880132 DOI: 10.1016/j.bbamcr.2025.119905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/09/2025] [Accepted: 01/17/2025] [Indexed: 01/31/2025]
Abstract
The imbalance of microglial homeostasis is highly associated with age-related neurological diseases, where cytosolic endogenous DNA is also likely to be found. As the main medium for storing biological information, endogenous DNA could be localized to cellular compartments normally free of DNA when cells are stimulated. However, the intracellular trafficking of endogenous DNA remains unidentified. In this study, we demonstrated that nuclear DNA (nDNA) and mitochondrial DNA (mtDNA), as the components of endogenous DNA, undergo different intracellular trafficking under conditions of microglial homeostasis imbalance induced by serum starvation. Upon detecting various components of endogenous DNA in the cytoplasmic and extracellular microglia, we found that cytosolic nDNA primarily exists in a free form and undergoes degradation through the autophagy-lysosome pathway. In contrast, cytosolic mtDNA predominantly exists in a membrane-wrapped form and is trafficked through both exosome and autophagy-lysosome pathways, with the exosome pathway serving as the primary one. When the autophagy-lysosome pathway was inhibited, there was an increase in exosomes. More importantly, the inhibition of the autophagy-lysosome pathway resulted in enhanced trafficking of mtDNA through the exosome pathway. These findings unveiled the crosstalk between these two pathways in the trafficking of microglial cytosolic DNA and thus provide new insights into intervening in age-related neurological diseases.
Collapse
Affiliation(s)
- Liyan Zhou
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zilong Wu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaoqing Yi
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Dongxue Xie
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jufen Wang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wenhe Wu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
33
|
Gonul CP, Kiser C, Yaka EC, Oz D, Hunerli D, Yerlikaya D, Olcum M, Keskinoglu P, Yener G, Genc S. Microglia-like cells from patient monocytes demonstrate increased phagocytic activity in probable Alzheimer's disease. Mol Cell Neurosci 2025; 132:103990. [PMID: 39732446 DOI: 10.1016/j.mcn.2024.103990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that is characterized by the accumulation of amyloid plaques, phosphorylated tau tangles and microglia toxicity, resulting in neuronal death and cognitive decline. Since microglia are recognized as one of the key players in the disease, it is crucial to understand how microglia operate in disease conditions and incorporate them into models. The studies on human microglia functions are thought to reflect the post-symptomatic stage of the disease. Recently developed methods involve induced microglia-like cells (iMGs) generated from patients' blood monocytes or induced pluripotent stem cells (iPSCs) as an alternative to studying the microglia cells in vitro. In this research, we aimed to investigate the phenotype and inflammatory responses of iMGs from AD patients. Monocytes derived from blood using density gradient centrifugation were differentiated into iMGs using a cytokine cocktail, including granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-34 (IL-34). After differentiation, cells were assessed by morphological analysis and a microglia surface marker, TMEM119. We used stimulants, lipopolysaccharide (LPS) and beta-amyloid, to examine iMGs' functions. Results showed that iMGs derived from AD patients exhibited increased secretion of pro-inflammatory cytokines upon LPS stimulation. Furthermore, their phagocytic ability was also heightened in stimulated and unstimulated conditions, with cells derived from patients showing increased phagocytic activity compared to healthy controls. Overall, these findings suggest that iMGs derived from patients using the direct conversion method possess characteristics of human microglia, making them an easy and promising model for studying microglia function in AD.
Collapse
Affiliation(s)
- Ceren Perihan Gonul
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Türkiye; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
| | - Cagla Kiser
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Türkiye; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
| | - Emis Cansu Yaka
- Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University, Izmir, Türkiye; Department of Neurology, Izmir City Hospital, Izmir, Türkiye
| | - Didem Oz
- Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University, Izmir, Türkiye; Department of Neurology, Dokuz Eylul University Hospital, Izmir, Türkiye; Global Brain Health Institute, University of California, San Francisco, USA
| | - Duygu Hunerli
- Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University, Izmir, Türkiye
| | - Deniz Yerlikaya
- Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University, Izmir, Türkiye
| | - Melis Olcum
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Türkiye
| | - Pembe Keskinoglu
- Department of Biostatistics and Medical Informatics, Basic Medical Sciences, Faculty of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Gorsev Yener
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Türkiye; Department of Neurology, Dokuz Eylul University Hospital, Izmir, Türkiye
| | - Sermin Genc
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Türkiye; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye; Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University, Izmir, Türkiye.
| |
Collapse
|
34
|
Bishnoi IR, Bordt EA. Sex and Region-Specific Differences in Microglial Morphology and Function Across Development. NEUROGLIA (BASEL, SWITZERLAND) 2025; 6:2. [PMID: 40181886 PMCID: PMC11967618 DOI: 10.3390/neuroglia6010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Microglia are exceptionally dynamic resident innate immune cells within the central nervous system, existing on a continuum of morphologies and functions throughout their lifespan. They play vital roles in response to injuries and infections, clearing cellular debris, and maintaining neural homeostasis throughout development. Emerging research suggests that microglia are strongly influenced by biological factors, including sex, developmental stage, and their local environment. This review synthesizes findings on sex differences in microglial morphology and function in key brain regions, including the frontal cortex, hippocampus, amygdala, hypothalamus, basal ganglia, and cerebellum, across the lifespan. Where available, we examine how gonadal hormones influence these microglial characteristics. Additionally, we highlight the limitations of relying solely on morphology to infer function and underscore the need for comprehensive, multimodal approaches to guide future research. Ultimately, this review aims to advance the dialogue on these spatiotemporally heterogeneous cells and their implications for sex differences in brain function and vulnerability to neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Indra R. Bishnoi
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Evan A. Bordt
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| |
Collapse
|
35
|
Matsuzaki H, Pan C, Komohara Y, Yamada R, Yano H, Fujiwara Y, Kai K, Mukasa A. The roles of glioma-associated macrophages/microglia and potential targets for anti-glioma therapy. Immunol Med 2025; 48:24-32. [PMID: 39391957 DOI: 10.1080/25785826.2024.2411035] [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: 05/22/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024] Open
Abstract
Glioblastoma (GBM) is the central nervous system tumor with the most aggressive behavior, and no definitive therapy has yet been found. The tumor microenvironment of GBM is immunosuppressive and is considered a 'cold tumor' with low lymphocytic infiltration, but is characterized by a high proportion of glioma-associated macrophages/microglia (GAMs). GAMs promote tumor growth and also affect treatment resistance in GBM. In this review, we describe the origin and classification of GAMs in humans and describe the mechanisms of their activation and the cell-cell interactions between tumor cells and GAMs. We also describe the history of GAM detection methods, especially immunohistochemistry, and discusses the merits and limitations of these techniques. In addition, we summarized chemotactic factors for GAMs and the therapies targeting these factors. Recent single-cell RNA analysis and spatial analysis add new insights to our previous knowledge of GAMs. Based on these studies, GBM therapies targeting GAMs are expected to be further developed.
Collapse
Affiliation(s)
- Hiroaki Matsuzaki
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Department of Neurosurgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Cheng Pan
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Rin Yamada
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Department of Diagnostic Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiromu Yano
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yukio Fujiwara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Keitaro Kai
- Department of Neurosurgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Akitake Mukasa
- Department of Neurosurgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
36
|
Liddelow SA, Zhang Y, Sloan SA. WebSEQ: A New Tool for Democratizing Omics Data Sharing. Glia 2025; 73:678-682. [PMID: 39722526 DOI: 10.1002/glia.24646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/24/2024] [Accepted: 11/04/2024] [Indexed: 12/28/2024]
Abstract
The relative ease of generation and proliferation of omics datasets has moved considerably faster than the effective dissemination of these data to the scientific community. Despite advancements in making raw data publicly available, many researchers struggle with data analysis and integration. We propose sharing analyzed data through user-friendly platforms to enhance accessibility. Here, we present a free, online tool, for sharing basic omics data in a searchable and user-friendly format. Importantly, it requires no coding or prior computational knowledge to build-only a data spreadsheet. Overall, this tool facilitates the exploration of transcriptomic, proteomic, and metabolomics data, which is crucial for understanding glial diversity and function. This initiative underscores the importance of accessible molecular data in advancing neuroscience research.
Collapse
Affiliation(s)
- Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York City, New York, USA
| | - Ye Zhang
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Steven A Sloan
- Department of Human Genetics, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
37
|
Wang D, Fukuda T, Wu T, Xu X, Isaji T, Gu J. Exogenous L-fucose attenuates depression induced by chronic unpredictable stress: Implicating core fucosylation has an antidepressant potential. J Biol Chem 2025; 301:108230. [PMID: 39864626 PMCID: PMC11879694 DOI: 10.1016/j.jbc.2025.108230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/14/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025] Open
Abstract
Core fucosylation is one of the most essential modifications of the N-glycans, catalyzed by α1,6-fucosyltransferase (Fut8), which transfers fucose from guanosine 5'-diphosphate (GDP)-fucose to the innermost N-acetylglucosamine residue of N-glycans in an α1-6 linkage. Our previous studies demonstrated that lipopolysaccharide (LPS) can induce a more robust neuroinflammatory response in Fut8 homozygous knockout (KO) (Fut8-/-) and heterozygous KO (Fut8+/-) mice contrasted to the wild-type (Fut8+/+) mice. Exogenous administration of L-fucose suppressed LPS-induced neuroinflammation. Numerous studies indicate that neuroinflammation plays a vital role in the development of depression. Here, we investigated whether core fucosylation regulates depression induced by chronic unpredictable stress (CUS), a well-established model for depression. Our results showed that Fut8+/- mice exhibited depressive-like behaviors and increased neuroinflammation earlier than Fut8+/+ mice. Administration of L-fucose significantly reduced CUS-induced depressive-like behaviors and pro-inflammatory cytokine levels in Fut8+/- mice. The L-fucose treatment produced antidepressant effects by attenuating the complex formation between gp130 and the interleukin-6 (IL-6) receptor and the JAK2/STAT3 signaling pathway. Notably, L-fucose treatment increased dendritic spine density and postsynaptic density protein 95 (PSD-95) expression, which were suppressed in CUS-induced depression. Furthermore, the effects of L-fucose on the CUS-induced depression were also observed in Fut8+/+ mice. Our results clearly demonstrate that L-fucose ameliorates neuroinflammation and synaptic defects in CUS-induced depression, implicating that core fucosylation has significant anti-neuroinflammatory activity and an antidepressant potential.
Collapse
Affiliation(s)
- Dan Wang
- Division of Regulatory Glycobiology, Graduate School of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Tomohiko Fukuda
- Division of Regulatory Glycobiology, Graduate School of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan; Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan.
| | - Tiangui Wu
- Division of Regulatory Glycobiology, Graduate School of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Xing Xu
- Division of Regulatory Glycobiology, Graduate School of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Tomoya Isaji
- Division of Regulatory Glycobiology, Graduate School of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan; Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Jianguo Gu
- Division of Regulatory Glycobiology, Graduate School of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan; Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan.
| |
Collapse
|
38
|
Zhu CC, Zheng YL, Gong C, Chen BL, Guo JB. Role of Exercise on Neuropathic Pain in Preclinical Models: Perspectives for Neuroglia. Mol Neurobiol 2025; 62:3684-3696. [PMID: 39316356 DOI: 10.1007/s12035-024-04511-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] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 09/15/2024] [Indexed: 09/25/2024]
Abstract
The benefits of exercise on neuropathic pain (NP) have been demonstrated in numerous studies. In recent studies, inflammation, neurotrophins, neurotransmitters, and endogenous opioids are considered as the main mechanisms. However, the role of exercise in alleviating NP remains unclear. Neuroglia, widely distributed in both the central and peripheral nervous systems, perform functions such as support, repair, immune response, and maintenance of normal neuronal activity. A large number of studies have shown that neuroglia play an important role in the occurrence and development of NP, and exercise can alleviate NP by regulating neuroglia. This article reviewed the involvement of neuroglia in the development of NP and their role in the exercise treatment of NP, intending to provide a theoretical basis for the exercise treatment strategy of NP.
Collapse
Affiliation(s)
- Chen-Chen Zhu
- The Second School of Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- School of Rehabilitation Medicine, Nanjing Medical University, Nanjing, China
| | - Yi-Li Zheng
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, 200438, China
| | - Chan Gong
- The Second School of Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- School of Rehabilitation Medicine, Nanjing Medical University, Nanjing, China
| | - Bing-Lin Chen
- The Second School of Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| | - Jia-Bao Guo
- The Second School of Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
39
|
Yano S, Asami N, Kishi Y, Takeda I, Kubotani H, Hattori Y, Kitazawa A, Hayashi K, Kubo KI, Saeki M, Maeda C, Hiraki C, Teruya RI, Taketomi T, Akiyama K, Okajima-Takahashi T, Sato B, Wake H, Gotoh Y, Nakajima K, Ichinohe T, Nagata T, Chiba T, Tsuruta F. Propagation of neuronal micronuclei regulates microglial characteristics. Nat Neurosci 2025; 28:487-498. [PMID: 39825140 DOI: 10.1038/s41593-024-01863-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/03/2024] [Indexed: 01/20/2025]
Abstract
Microglia-resident immune cells in the central nervous system-undergo morphological and functional changes in response to signals from the local environment and mature into various homeostatic states. However, niche signals underlying microglial differentiation and maturation remain unknown. Here, we show that neuronal micronuclei (MN) transfer to microglia, which is followed by changing microglial characteristics during the postnatal period. Neurons passing through a dense region of the developing neocortex give rise to MN and release them into the extracellular space, before being incorporated into microglia and inducing morphological changes. Two-photon imaging analyses have revealed that microglia incorporating MN tend to slowly retract their processes. Loss of the cGAS gene alleviates effects on micronucleus-dependent morphological changes. Neuronal MN-harboring microglia also exhibit unique transcriptome signatures. These results demonstrate that neuronal MN serve as niche signals that transform microglia, and provide a potential mechanism for regulation of microglial characteristics in the early postnatal neocortex.
Collapse
Affiliation(s)
- Sarasa Yano
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
- Chugai Life Science Park Yokohama, Chugai Pharmaceutical Co. Ltd., Yokohama, Japan
| | - Natsu Asami
- Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Japan
| | - Yusuke Kishi
- Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Ikuko Takeda
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, Myodaiji Okazaki, Japan
| | - Hikari Kubotani
- Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Japan
| | - Yuki Hattori
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ayako Kitazawa
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
- Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan
| | - Kanehiro Hayashi
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Ken-Ichiro Kubo
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
- Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan
| | - Mai Saeki
- Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Chihiro Maeda
- Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Japan
| | - Chihiro Hiraki
- Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Japan
| | - Rin-Ichiro Teruya
- Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Japan
| | - Takumi Taketomi
- School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Japan
| | - Kaito Akiyama
- College of Biological Sciences, School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | | | - Ban Sato
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
- Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Hiroaki Wake
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, Myodaiji Okazaki, Japan
- Department of Physiological Sciences, Graduate University for Advanced Studies SOKENDAI, Hayama, Japan
- Department of Systems Science, Center of Optical Scattering Image Science, Kobe University, Kobe, Japan
| | - Yukiko Gotoh
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Tokyo, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Takeshi Ichinohe
- Department of Infectious Disease Control, International Research Center for Infectious Diseases, Institute of Medical Science, The University of Tokyo Minato-ku, Tokyo, Japan
| | - Takeshi Nagata
- School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Japan
- Information and Communication Research Division, Mizuho Research and Technologies Ltd., Tokyo, Japan
- Faculty of Mathematical Informatics, Meiji Gakuin University, Yokohama, Japan
| | - Tomoki Chiba
- School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Japan
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Fuminori Tsuruta
- School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Japan.
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan.
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan.
- Center for Quantum and Information Life Sciences, University of Tsukuba, Tsukuba, Japan.
| |
Collapse
|
40
|
Tang C, Zhou QQ, Huang XF, Ju YY, Rao BL, Liu ZC, Jia YA, Bai ZP, Lin QY, Liu L, Qu J, Zhang J, Gao ML. Integration and functionality of human iPSC-derived microglia in a chimeric mouse retinal model. J Neuroinflammation 2025; 22:53. [PMID: 40016767 PMCID: PMC11869422 DOI: 10.1186/s12974-025-03393-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/20/2025] [Indexed: 03/01/2025] Open
Abstract
INTRODUCTION Microglia, the resident immune cells of the central nervous system, play a pivotal role in maintaining homeostasis, responding to injury, and modulating neuroinflammation. However, the limitations of rodent models in accurately representing human microglia have posed significant challenges in the study of retinal diseases. METHODS PLX5622 was used to eliminate endogenous microglia in mice through oral and intraperitoneal administration, followed by transplantation of human induced pluripotent stem cell-derived microglia (hiPSC-microglia, iMG) into retinal explants to create a novel ex vivo chimeric model containing xenotransplanted microglia (xMG). The number and proportion of xMG in the retina were quantified using retinal flat-mounting and immunostaining. To evaluate the proliferative capacity and synaptic pruning ability of xMG, the expression of Ki-67 and the phagocytosis of synaptic proteins SV2 and PSD95 was assessed. The chimeric model was stimulated with LPS, and single-cell RNA sequencing (scRNA-seq) was used to analyze transcriptomic changes in iMG and xMG. Mouse IL-34 antibody neutralization experiments were performed, and the behavior of xMG in retinal degenerative Pde6b-/- mice was examined. RESULTS We demonstrated that xenotransplanted microglia (xMG) successfully migrated to and localized within the mouse retina, adopting homeostatic morphologies. Our approach achieved over 86% integration of human microglia, which maintained key functions including proliferation, immune responsiveness, and synaptic pruning over a 14-day culture period. scRNA-seq of xMG revealed a shift in microglial signatures compared to monoculture iMG, indicating a transition to a more in vivo-like phenotype. In retinal degenerative Pde6b-/- mice, xMG exhibited activation and migrated toward degenerated photoreceptors. CONCLUSION This model provides a powerful platform for studying human microglia in the retinal context, offering significant insights for advancing research into retinal degenerative diseases and developing potential therapeutic strategies. Future applications of this model include using patient-derived iPSCs to investigate disease-specific microglial behaviors, thereby enhancing our understanding of microglia-related pathogenesis.
Collapse
Affiliation(s)
- Chun Tang
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Qi-Qi Zhou
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiu-Feng Huang
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Ya-Yi Ju
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Bi-Lin Rao
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhi-Cong Liu
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yi-An Jia
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhan-Pei Bai
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Qing-Yang Lin
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Lin Liu
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jia Qu
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China.
- The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, 325027, China.
| | - Jun Zhang
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China.
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China.
- Lead Contact, Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| | - Mei-Ling Gao
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, 325027, China.
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, China.
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| |
Collapse
|
41
|
Kumaraguru S, Morgan J, Wong FK. Activity-dependent regulation of microglia numbers by pyramidal cells during development shape cortical functions. SCIENCE ADVANCES 2025; 11:eadq5842. [PMID: 39970202 PMCID: PMC11838000 DOI: 10.1126/sciadv.adq5842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 01/15/2025] [Indexed: 02/21/2025]
Abstract
Beyond their role as immune sentinels, microglia are actively involved in establishing and maintaining cortical circuits. Alteration in microglial numbers has been associated with abnormal behaviors akin to those observed in neurodevelopmental disorders. Consequently, establishing the appropriate microglial numbers during development is crucial for ensuring normal cortical function. Here, we uncovered a dynamic relationship between pyramidal cells and microglia that tunes microglial numbers and development through distinct phases of mouse postnatal development. Changes in pyramidal cell activity during development induce differential release of activity-dependent proteins such as Activin A, which, in turn, adjusts microglial numbers accordingly. Decoupling of this relationship not only changes microglial numbers but has a long-term consequence on their role as synaptic organizers, which ultimately affects cortical function. Our findings reveal that microglia adapt their numbers to changes in pyramidal cell activity during a critical time window in development, consequently adjusting their numbers and function to the demands of the developing local circuits.
Collapse
Affiliation(s)
- Sanjana Kumaraguru
- Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - James Morgan
- Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Fong Kuan Wong
- Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, UK
| |
Collapse
|
42
|
Perochon T, Krsnik Z, Massimo M, Ruchiy Y, Romero AL, Mohammadi E, Li X, Long KR, Parkkinen L, Blomgren K, Lagache T, Menassa DA, Holcman D. Unraveling microglial spatial organization in the developing human brain with DeepCellMap, a deep learning approach coupled with spatial statistics. Nat Commun 2025; 16:1577. [PMID: 39948387 PMCID: PMC11825940 DOI: 10.1038/s41467-025-56560-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Mapping cellular organization in the developing brain presents significant challenges due to the multidimensional nature of the data, characterized by complex spatial patterns that are difficult to interpret without high-throughput tools. Here, we present DeepCellMap, a deep-learning-assisted tool that integrates multi-scale image processing with advanced spatial and clustering statistics. This pipeline is designed to map microglial organization during normal and pathological brain development and has the potential to be adapted to any cell type. Using DeepCellMap, we capture the morphological diversity of microglia, identify strong coupling between proliferative and phagocytic phenotypes, and show that distinct spatial clusters rarely overlap as human brain development progresses. Additionally, we uncover an association between microglia and blood vessels in fetal brains exposed to maternal SARS-CoV-2. These findings offer insights into whether various microglial phenotypes form networks in the developing brain to occupy space, and in conditions involving haemorrhages, whether microglia respond to, or influence changes in blood vessel integrity. DeepCellMap is available as an open-source software and is a powerful tool for extracting spatial statistics and analyzing cellular organization in large tissue sections, accommodating various imaging modalities. This platform opens new avenues for studying brain development and related pathologies.
Collapse
Affiliation(s)
- Theo Perochon
- Group of Data Modeling and Computational Biology, IBENS, École Normale Supérieure, Paris, France
| | - Zeljka Krsnik
- Croatian Institute for Brain Research, University of Zagreb, Zagreb, Croatia
| | - Marco Massimo
- Centre for Developmental Neurobiology, MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Yana Ruchiy
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | | | - Elyas Mohammadi
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Xiaofei Li
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Katherine R Long
- Centre for Developmental Neurobiology, MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Laura Parkkinen
- Department of Neuropathology and The Queen's College, University of Oxford, Oxford, United Kingdom
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Thibault Lagache
- BioImage Analysis Unit, CNRS UMR3691, Institut Pasteur, Université Paris Cité, Paris, France.
| | - David A Menassa
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden.
- Department of Neuropathology and The Queen's College, University of Oxford, Oxford, United Kingdom.
| | - David Holcman
- Group of Data Modeling and Computational Biology, IBENS, École Normale Supérieure, Paris, France.
- DAMPT, University of Cambridge, DAMPT and Churchill College, Cambridge, United Kingdom.
| |
Collapse
|
43
|
Kasper M, Karlstetter M, Wildschütz L, Scholz R, Busch M, Bauer D, Meyer Zu Hörste G, Thanos S, Langmann T, Heiligenhaus A. Kinetic changes in microglia-related retinal transcripts in experimental autoimmune uveoretinitis (EAU) of B10.RIII mice. J Neuroinflammation 2025; 22:37. [PMID: 39930455 PMCID: PMC11812248 DOI: 10.1186/s12974-025-03358-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 01/26/2025] [Indexed: 02/13/2025] Open
Abstract
In this study the retinal transcriptome was investigated during the development of experimental autoimmune uveoretinitis (EAU) in mice. EAU was induced by immunizing B10.RIII mice with human interphotoreceptor retinoid binding protein (hIRBP) 161-180 peptide. Genome-wide transcriptional profiles of EAU (day 7, 14 or 21 after immunization) and of control retinas were generated using DNA-microarrays and bioinformatic data mining. Microglia-associated transcripts were identified. Quantitative real-time polymerase chain reaction was performed to validate the expression of differentially expressed genes. Retinal transcript validation revealed that complement and interferon-related pathways, as well as gene clusters specific for antigen-processing and -presentation, and immunosuppression are involved during the course of the disease. Immunofluorescence analysis confirm that upregulated transcripts in EAU are also expressed by retinal microglia. Furthermore, the heterogenous expression patterns observed in retinal microglia, suggests the presence of different subpopulations of retinal microglia in EAU. This study expands our knowledge of the local immune processes involved in EAU pathology.
Collapse
Affiliation(s)
- Maren Kasper
- Department of Ophthalmology at St. Franziskus Hospital, Ophtha-Lab, Hohenzollernring 74, 48145, Münster, Germany.
| | - Marcus Karlstetter
- Chair of Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - Lena Wildschütz
- Department of Ophthalmology at St. Franziskus Hospital, Ophtha-Lab, Hohenzollernring 74, 48145, Münster, Germany
| | - Rebecca Scholz
- Chair of Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - Martin Busch
- Department of Ophthalmology at St. Franziskus Hospital, Ophtha-Lab, Hohenzollernring 74, 48145, Münster, Germany
| | - Dirk Bauer
- Department of Ophthalmology at St. Franziskus Hospital, Ophtha-Lab, Hohenzollernring 74, 48145, Münster, Germany
| | | | - Solon Thanos
- Institute for Experimental Ophthalmology, Westfalian Wilhelms-University of Münster, Münster, Germany
| | - Thomas Langmann
- Chair of Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - Arnd Heiligenhaus
- Department of Ophthalmology at St. Franziskus Hospital, Ophtha-Lab, Hohenzollernring 74, 48145, Münster, Germany
- University of Duisburg-Essen, Duisburg, Germany
| |
Collapse
|
44
|
Richardson TE, Orr ME, Orr TC, Rohde SK, Ehrenberg AJ, Thorn EL, Christie TD, Flores‐Almazan V, Afzal R, De Sanctis C, Maldonado‐Díaz C, Hiya S, Canbeldek L, Kulumani Mahadevan LS, Slocum C, Samanamud J, Clare K, Scibetta N, Yokoda RT, Koenigsberg D, Marx GA, Kauffman J, Goldstein A, Selmanovic E, Drummond E, Wisniewski T, White CL, Goate AM, Crary JF, Farrell K, Alosco ML, Mez J, McKee AC, Stein TD, Bieniek KF, Kautz TF, Daoud EV, Walker JM. Spatial proteomic differences in chronic traumatic encephalopathy, Alzheimer's disease, and primary age-related tauopathy hippocampi. Alzheimers Dement 2025; 21:e14487. [PMID: 39737785 PMCID: PMC11848160 DOI: 10.1002/alz.14487] [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: 08/17/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 01/01/2025]
Abstract
INTRODUCTION Alzheimer's disease (AD), primary age-related tauopathy (PART), and chronic traumatic encephalopathy (CTE) all feature hyperphosphorylated tau (p-tau)-immunoreactive neurofibrillary degeneration, but differ in neuroanatomical distribution and progression of neurofibrillary degeneration and amyloid beta (Aβ) deposition. METHODS We used Nanostring GeoMx Digital Spatial Profiling to compare the expression of 70 proteins in neurofibrillary tangle (NFT)-bearing and non-NFT-bearing neurons in hippocampal CA1, CA2, and CA4 subregions and entorhinal cortex of cases with autopsy-confirmed AD (n = 8), PART (n = 7), and CTE (n = 5). RESULTS There were numerous subregion-specific differences related to Aβ processing, autophagy/proteostasis, inflammation, gliosis, oxidative stress, neuronal/synaptic integrity, and p-tau epitopes among these different disorders. DISCUSSION These results suggest that there are subregion-specific proteomic differences among the neurons of these disorders, which appear to be influenced to a large degree by the presence of hippocampal Aβ. These proteomic differences may play a role in the differing hippocampal p-tau distribution and pathogenesis of these disorders. HIGHLIGHTS Alzheimer's disease neuropathologic change (ADNC), possible primary age-related tauopathy (PART), definite PART, and chronic traumatic encephalopathy (CTE) can be differentiated based on the proteomic composition of their neurofibrillary tangle (NFT)- and non-NFT-bearing neurons. The proteome of these NFT- and non-NFT-bearing neurons is largely correlated with the presence or absence of amyloid beta (Aβ). Neurons in CTE and definite PART (Aβ-independent pathologies) share numerous proteomic similarities that distinguish them from ADNC and possible PART (Aβ-positive pathologies).
Collapse
Affiliation(s)
- Timothy E. Richardson
- Department of PathologyMolecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Miranda E. Orr
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
- St. Louis VA Medical CenterSt. LouisMissouriUSA
| | - Timothy C. Orr
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Susan K. Rohde
- Department of PathologyMolecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of PathologyVrije Universiteit AmsterdamAmsterdamthe Netherlands
- Department of NeuroscienceVrije Universiteit AmsterdamAmsterdamthe Netherlands
- Department of Human GeneticsGenomics of Neurodegenerative Diseases and AgingVrije Universiteit AmsterdamAmsterdamthe Netherlands
- Department of NeurologyAlzheimer Center AmsterdamNeuroscienceVrije Universiteit AmsterdamAmsterdamthe Netherlands
| | - Alexander J. Ehrenberg
- Memory and Aging CenterWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Helen Wills Neuroscience InstituteUniversity of CaliforniaBerkeleyCaliforniaUSA
- Innovative Genomics InstituteUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Emma L. Thorn
- Department of PathologyMolecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Neuropathology Brain Bank & Research CoREIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Thomas D. Christie
- Department of PathologyMolecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Neuropathology Brain Bank & Research CoREIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Victoria Flores‐Almazan
- Department of PathologyMolecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Neuropathology Brain Bank & Research CoREIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Robina Afzal
- Department of PathologyMolecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Neuropathology Brain Bank & Research CoREIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Claudia De Sanctis
- Department of PathologyMolecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Neuropathology Brain Bank & Research CoREIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Carolina Maldonado‐Díaz
- Department of PathologyMolecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Satomi Hiya
- Department of PathologyMolecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Leyla Canbeldek
- Department of PathologyMolecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | | | - Cheyanne Slocum
- Department of PathologyMolecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Jorge Samanamud
- Department of PathologyMolecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Kevin Clare
- Department of PathologyMolecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Nicholas Scibetta
- Department of PathologyMolecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Raquel T. Yokoda
- Department of PathologyAlbert Einstein College of MedicineMontefiore Medical CenterBronxNew YorkUSA
| | - Daniel Koenigsberg
- Department of PathologyMolecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Neuropathology Brain Bank & Research CoREIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Nash Family Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Artificial Intelligence & Human HealthIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Ronald M. Loeb Center for Alzheimer's DiseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Friedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Gabriel A. Marx
- Department of PathologyMolecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Neuropathology Brain Bank & Research CoREIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Nash Family Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Artificial Intelligence & Human HealthIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Ronald M. Loeb Center for Alzheimer's DiseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Friedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of NeurologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Justin Kauffman
- Department of PathologyMolecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Neuropathology Brain Bank & Research CoREIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Nash Family Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Artificial Intelligence & Human HealthIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Ronald M. Loeb Center for Alzheimer's DiseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Friedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Adam Goldstein
- Department of PathologyMolecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Neuropathology Brain Bank & Research CoREIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Enna Selmanovic
- Nash Family Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Friedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Eleanor Drummond
- Brain & Mind Center and School of Medical SciencesFaculty of Medicine and HealthUniversity of SydneyCamperdownNew South WalesAustralia
| | - Thomas Wisniewski
- Department of PathologyNew York University Grossman School of MedicineNew YorkNew YorkUSA
- Department of PsychiatryNew York University Grossman School of MedicineNew YorkNew YorkUSA
- Center for Cognitive NeurologyDepartment of NeurologyNew York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Charles L. White
- Department of PathologyUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Alison M. Goate
- Nash Family Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Ronald M. Loeb Center for Alzheimer's DiseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - John F. Crary
- Department of PathologyMolecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Neuropathology Brain Bank & Research CoREIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Nash Family Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Artificial Intelligence & Human HealthIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Ronald M. Loeb Center for Alzheimer's DiseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Friedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Kurt Farrell
- Department of PathologyMolecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Neuropathology Brain Bank & Research CoREIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Nash Family Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Artificial Intelligence & Human HealthIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Ronald M. Loeb Center for Alzheimer's DiseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Friedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Michael L. Alosco
- Department of NeurologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Boston University Alzheimer's Disease Research Center and BU CTE CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Jesse Mez
- Department of NeurologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Boston University Alzheimer's Disease Research Center and BU CTE CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Ann C. McKee
- Department of NeurologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Boston University Alzheimer's Disease Research Center and BU CTE CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- VA Boston Healthcare SystemBostonMassachusettsUSA
- VA Bedford Healthcare SystemBedfordMassachusettsUSA
| | - Thor D. Stein
- Department of NeurologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Boston University Alzheimer's Disease Research Center and BU CTE CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- VA Boston Healthcare SystemBostonMassachusettsUSA
- VA Bedford Healthcare SystemBedfordMassachusettsUSA
| | - Kevin F. Bieniek
- Department of Pathology & Laboratory MedicineUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Tiffany F. Kautz
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Elena V. Daoud
- Department of PathologyUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Jamie M. Walker
- Department of PathologyMolecular, and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Neuropathology Brain Bank & Research CoREIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Nash Family Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| |
Collapse
|
45
|
Kang M, Nasrollahi A, Cheng F, Yao Y. Screening and Identification of Brain Pericyte-Selective Markers. CNS Neurosci Ther 2025; 31:e70247. [PMID: 39912338 PMCID: PMC11799917 DOI: 10.1111/cns.70247] [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: 12/03/2024] [Revised: 01/10/2025] [Accepted: 01/19/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Pericytes, a type of mural cells, exert important functions in the CNS. One major challenge in pericyte research is the lack of pericyte-specific and subpopulation-specific markers. METHODS To address this knowledge gap, we first generated a novel transgenic mouse line in which vascular smooth muscle cells (vSMCs) are permanently labeled with tdTomato. Next, we isolated PDGFRβ+tdTomato- pericytes and PDGFRβ+tdTomato+ vSMCs from the brains of these mice and subsequently performed RNAseq analysis to identify pericyte-enriched genes. RESULTS Using this approach, we successfully identified 40 pericyte-enriched genes and 158 vSMC-enriched genes, which are involved in different biological processes and molecular functions. Using ISH/IHC analysis, we found that Pla1a and Cox4i2 were predominantly enriched in subpopulations of brain pericytes, although they also marked some non-vascular parenchymal cells. CONCLUSIONS These findings suggest that Pla1a and Cox4i2 preferably label subpopulations of pericytes in the brain compared to vSMCs, and thus, they may be useful in distinguishing these populations.
Collapse
Affiliation(s)
- Minkyung Kang
- Department of Molecular Pharmacology and Physiology, Morsani College of MedicineUniversity of South FloridaTampaFloridaUSA
| | - Ava Nasrollahi
- Department of Molecular Pharmacology and Physiology, Morsani College of MedicineUniversity of South FloridaTampaFloridaUSA
| | - Feng Cheng
- Department of Pharmaceutical Science, College of PharmacyUniversity of South FloridaTampaFloridaUSA
| | - Yao Yao
- Department of Molecular Pharmacology and Physiology, Morsani College of MedicineUniversity of South FloridaTampaFloridaUSA
| |
Collapse
|
46
|
Kim HG, Berdasco C, Nairn AC, Kim Y. The WAVE complex in developmental and adulthood brain disorders. Exp Mol Med 2025; 57:13-29. [PMID: 39774290 PMCID: PMC11799376 DOI: 10.1038/s12276-024-01386-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 10/09/2024] [Accepted: 10/31/2024] [Indexed: 01/11/2025] Open
Abstract
Actin polymerization and depolymerization are fundamental cellular processes required not only for the embryonic and postnatal development of the brain but also for the maintenance of neuronal plasticity and survival in the adult and aging brain. The orchestrated organization of actin filaments is controlled by various actin regulatory proteins. Wiskott‒Aldrich syndrome protein-family verprolin-homologous protein (WAVE) members are key activators of ARP2/3 complex-mediated actin polymerization. WAVE proteins exist as heteropentameric complexes together with regulatory proteins, including CYFIP, NCKAP, ABI and BRK1. The activity of the WAVE complex is tightly regulated by extracellular cues and intracellular signaling to execute its roles in specific intracellular events in brain cells. Notably, dysregulation of the WAVE complex and WAVE complex-mediated cellular processes confers vulnerability to a variety of brain disorders. De novo mutations in WAVE genes and other components of the WAVE complex have been identified in patients with developmental disorders such as intellectual disability, epileptic seizures, schizophrenia, and/or autism spectrum disorder. In addition, alterations in the WAVE complex are implicated in the pathophysiology of Alzheimer's disease and Parkinson's disease, as well as in behavioral adaptations to psychostimulants or maladaptive feeding.
Collapse
Affiliation(s)
- Hyung-Goo Kim
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, 08854, USA
| | - Clara Berdasco
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, 08854, USA
| | - Angus C Nairn
- Department of Psychiatry, Yale School of Medicine, Connecticut Mental Health Center, New Haven, CT, USA
| | - Yong Kim
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, 08854, USA.
- Brain Health Institute, Rutgers University, Piscataway, NJ, 08854, USA.
| |
Collapse
|
47
|
Hamagami N, Kapadia D, Abduljawad N, Cheng Z, McLaughlin L, Singhania D, Barclay KM, Yang J, Sun Z, Bayguinov P, Yu G, Gabel HW, Li Q. Microglial plasticity governed by state-specific enhancer landscapes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635595. [PMID: 39975390 PMCID: PMC11838276 DOI: 10.1101/2025.01.30.635595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Single-cell transcriptomic studies have identified distinct microglial subpopulations with shared and divergent gene signatures across development, aging and disease. Whether these microglial subsets represent ontogenically separate lineages of cells, or they are manifestations of plastic changes of microglial states downstream of some converging signals is unknown. Furthermore, despite the well-established role of enhancer landscapes underlying the identity of microglia, to what extent histone modifications and DNA methylation regulate microglial state switches at enhancers have not been defined. Here, using genetic fate mapping, we demonstrate the common embryonic origin of proliferative-region-associated microglia (PAM) enriched in developing white matter, and track their dynamic transitions into disease-associated microglia (DAM) and white matter-associated microglia (WAM) states in disease and aging contexts, respectively. This study links spatiotemporally discrete microglial states through their transcriptomic and epigenomic plasticity, while revealing state-specific histone modification profiles that govern state switches in health and disease.
Collapse
Affiliation(s)
- Nicole Hamagami
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO 63110, USA
- These authors contributed equally
| | - Dvita Kapadia
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- These authors contributed equally
| | - Nora Abduljawad
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Neuroscience Graduate Program, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Zuolin Cheng
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Liam McLaughlin
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Darsh Singhania
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Kia M. Barclay
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Neuroscience Graduate Program, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jin Yang
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Zhixin Sun
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Peter Bayguinov
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Guoqiang Yu
- Department of Automation, Tsinghua University, Beijing 100084, China
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| | - Harrison W. Gabel
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- These authors contributed equally
| | - Qingyun Li
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Department of Genetics, Washington University School of Medicine in St. Louis, School of Medicine, St. Louis, MO 63110, USA
- These authors contributed equally
- Lead contact
| |
Collapse
|
48
|
Schepanski S, Ngoumou GB, Buss C, Seifert G. Assessing in-vitro models for microglial development and fetal programming: a critical review. Front Immunol 2025; 16:1538920. [PMID: 39944696 PMCID: PMC11814449 DOI: 10.3389/fimmu.2025.1538920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/08/2025] [Indexed: 05/09/2025] Open
Abstract
This review evaluates in-vitro models for studying how maternal influences during pregnancy impact the development of offspring microglia, the immune cells of the central nervous system. The models examined include primary microglia cultures, microglia cell lines, iPSC-derived microglia, PBMC-induced microglia-like cells, 3D brain organoids derived from iPSCs, and Hofbauer cells. Each model is assessed for its ability to replicate the in-vivo environment of the developing brain, with a focus on their strengths, limitations, and practical challenges. Key factors such as scalability, genetic and epigenetic fidelity, and physiological relevance are highlighted. Microglia cell lines are highly scalable but lack genetic and epigenetic fidelity. iPSC-derived microglia provide moderate physiological relevance and patient-specific genetic insights but face operational and epigenetic challenges inherent to reprogramming. 3D brain organoids, derived from iPSCs, offer an advanced platform for studying complex neurodevelopmental processes but require extensive resources and technical expertise. Hofbauer cells, which are fetal macrophages located in the placenta and share a common developmental origin with microglia, are uniquely exposed to prenatal maternal factors and, depending on fetal barrier maturation, exhibit variable epigenetic fidelity. This makes them particularly useful for exploring the impact of maternal influences on fetal programming of microglial development. The review concludes that no single model comprehensively captures all aspects of maternal influences on microglial development, but it offers guidance on selecting the most appropriate model based on specific research objectives and experimental constraints.
Collapse
Affiliation(s)
- Steven Schepanski
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité Competence Center for Traditional and Integrative Medicine (CCCTIM), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Pediatrics, Division of Oncology and Hematology, Berlin, Germany
| | - Gonza B. Ngoumou
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité Competence Center for Traditional and Integrative Medicine (CCCTIM), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Pediatrics, Division of Oncology and Hematology, Berlin, Germany
| | - Claudia Buss
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Medical Psychology, Berlin, Germany
- University of California, Irvine, Development, Health and Disease Research Program, Irvine, CA, United States
- German Center for Child and Adolescent Health (DZKJ), Partner Site Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Mental Health (DZPG), Partner Site Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Georg Seifert
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité Competence Center for Traditional and Integrative Medicine (CCCTIM), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Pediatrics, Division of Oncology and Hematology, Berlin, Germany
| |
Collapse
|
49
|
Le LHD, Eliseeva S, Plunk E, Kara-Pabani K, Li H, Yarovinsky F, Majewska AK. The microglial response to inhibition of Colony-stimulating-factor-1 receptor by PLX3397 differs by sex in adult mice. Cell Rep 2025; 44:115176. [PMID: 39842435 PMCID: PMC11877653 DOI: 10.1016/j.celrep.2024.115176] [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: 04/08/2024] [Revised: 10/08/2024] [Accepted: 12/18/2024] [Indexed: 01/24/2025] Open
Abstract
Microglia, the resident macrophages of the brain, are derived from the yolk sac and colonize the brain before the blood-brain barrier forms. Once established, they expand locally and require Colony-stimulating-factor-1 receptor (CSF1R) signaling for their development and maintenance. CSF1R inhibitors have been used extensively to deplete microglia in the healthy and diseased brain. In this study, we demonstrated sex-dependent differences in the microglial response to the CSF1R inhibitor PLX3397. Male mice exhibited greater microglial depletion compared to females. Transcriptomic and flow cytometry analysis revealed sex-specific differences in the remaining microglia population, with female microglia upregulating autophagy and proteostasis pathways while male microglia increased mitobiogenesis. Furthermore, manipulating key microglial receptors by using different transgenic mouse lines resulted in changes in depletion efficacies that were also sex dependent. These findings suggest sex-dependent microglial survival mechanisms, which might contribute to the well-documented sex differences in various neurological disorders.
Collapse
Affiliation(s)
- Linh H D Le
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY 14642, USA
| | - Sophia Eliseeva
- Department of Microbiology and Immunology, Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | - Elizabeth Plunk
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY 14642, USA; Department of Environmental Medicine, University of Rochester, Rochester, NY 14642, USA
| | - Kallam Kara-Pabani
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY 14642, USA
| | - Herman Li
- Medical Scientist Training Program, University of Rochester, Rochester, NY 14642, USA
| | - Felix Yarovinsky
- Department of Microbiology and Immunology, Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | - Ania K Majewska
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY 14642, USA; Center for Visual Science, University of Rochester, Rochester, NY 14642, USA.
| |
Collapse
|
50
|
Falk I, Maric D, Leibovitch E, Sati P, Lefeuvre J, Luciano NJ, Guy J, Ha SK, Owen DR, Aigbirhio F, Matthews PM, Reich DS, Jacobson S. Characteristics of TSPO expression in marmoset EAE. J Neuroinflammation 2025; 22:19. [PMID: 39871344 PMCID: PMC11773908 DOI: 10.1186/s12974-025-03343-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/10/2025] [Indexed: 01/29/2025] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) and is a leading non-traumatic cause of disability in young adults. The 18 kDa Translocator Protein (TSPO) is a mitochondrial protein and positron emission tomography (PET)-imaging target that is highly expressed in MS brain lesions. It is used as an inflammatory biomarker and has been proposed as a therapeutic target. However, its specific pathological significance in humans is not well understood. Experimental autoimmune encephalomyelitis (EAE) in the common marmoset is a well-established primate model of MS. Studying TSPO expression in this model will enhance our understanding of its expression in MS. This study therefore characterizes patterns of TSPO expression in fixed CNS tissues from one non-EAE control marmoset and 8 EAE marmosets using multiplex immunofluorescence. In control CNS tissue, we find that TSPO is expressed in the leptomeninges, ependyma, and over two-thirds of Iba1 + microglia, but not astrocytes or neurons. In Iba1 + cells in both control and acute EAE tissue, we find that TSPO is co-expressed with markers of antigen presentation (CD74), early activation (MRP14), phagocytosis (CD163) and anti-inflammatory phenotype (Arg1); a high level of TSPO expression is not restricted to a particular microglial phenotype. While TSPO is expressed in over 88% of activated Iba1 + cells in acute lesions in marmoset EAE, it also is sometimes observed in subsets of astrocytes and neurons. Additionally, we find the percentage of Iba1 + cells expressing TSPO declines significantly in lesions > 5 months old and may be as low as 13% in chronic lesions. However, we also find increased astrocytic TSPO expression in chronic-appearing lesions with astrogliosis. Finally, we find expression of TSPO in a subset of neurons, most frequently GLS2 + glutamatergic neurons. The shift in TSPO expression from Iba + microglia/macrophages to astrocytes over time is similar to patterns suggested by earlier neuropathology studies in MS. Thus, marmoset EAE appears to be a clinically relevant model for the study of TSPO in immune dysregulation in human disease.
Collapse
Affiliation(s)
- Irene Falk
- Viral Immunology Section, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Building 10, Room 5C103, 10 Center Drive, Bethesda, MD, 20892-1400, USA
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Emily Leibovitch
- Viral Immunology Section, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Building 10, Room 5C103, 10 Center Drive, Bethesda, MD, 20892-1400, USA
| | - Pascal Sati
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer Lefeuvre
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Nicholas J Luciano
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Joseph Guy
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Seung-Kwon Ha
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - David R Owen
- Department of Brain Sciences, Imperial College London, London, UK
| | - Franklin Aigbirhio
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Steven Jacobson
- Viral Immunology Section, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Building 10, Room 5C103, 10 Center Drive, Bethesda, MD, 20892-1400, USA.
| |
Collapse
|