1
|
Frosch M, Prinz M. Niche-specific therapeutic targeting of myeloid cells in the central nervous system. Immunity 2025; 58:1101-1119. [PMID: 40324377 DOI: 10.1016/j.immuni.2025.03.016] [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: 01/15/2025] [Revised: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 05/07/2025]
Abstract
The central nervous system (CNS) can be subdivided into distinct anatomical and functional compartments, including the parenchyma, perivascular space, leptomeninges, and dura mater, etc. Each compartment hosts distinct immune cell populations, such as monocytes and diverse macrophages, which play critical roles in local tissue homeostasis and regional disease pathogenesis. Advances in single-cell technologies have revealed complex immune cell compositions and functions in these anatomical regions. This review summarizes the latest approaches for modulating myeloid cell subsets in a compartment-specific manner, including cellular strategies such as stem cell therapy, ex vivo gene treatment, bone marrow transplantation, as well as non-cellular strategies like antibodies, small molecules, and viral gene delivery to augment CNS immune responses and improve disease outcomes. We also discuss the challenges and requirements of translating targeting strategies from mice to humans.
Collapse
Affiliation(s)
- Maximilian Frosch
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany; CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
2
|
Bastos J, O'Brien C, Vara-Pérez M, Mampay M, van Olst L, Barry-Carroll L, Kancheva D, Leduc S, Lievens AL, Ali L, Vlasov V, Meysman L, Shakeri H, Roelandt R, Van Hove H, De Vlaminck K, Scheyltjens I, Yaqoob F, Lombroso SI, Breugelmans M, Faron G, Gomez-Nicola D, Gate D, Bennett FC, Movahedi K. Monocytes can efficiently replace all brain macrophages and fetal liver monocytes can generate bona fide SALL1 + microglia. Immunity 2025; 58:1269-1288.e12. [PMID: 40311613 DOI: 10.1016/j.immuni.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 01/29/2025] [Accepted: 04/07/2025] [Indexed: 05/03/2025]
Abstract
Microglia and border-associated macrophages (BAMs) are critical for brain health, and their dysfunction is associated to disease. Replacing brain macrophages holds substantial therapeutic promise but remains challenging. Here, we demonstrate that monocytes can efficiently replace all brain macrophages. Monocytes readily replaced embryonal BAMs upon their depletion and engrafted as monocyte-derived microglia (Mo-Microglia) upon more sustained niche availability. Mo-Microglia expanded comparably to their embryonic counterparts and showed similar longevity. However, monocytes were unable to replicate the distinct identity of embryonically derived BAMs and microglia. Using xenotransplantation, we found that human monocytes exhibited similar behavior, enabling identification of putative Mo-Microglia in Alzheimer's disease individuals. In mice and humans, monocyte ontogeny shaped their identity as brain macrophages. Importantly, mouse fetal liver monocytes exhibited a distinct epigenetic landscape and could develop a bona fide microglial identity. Our results illuminate brain macrophage development and highlight monocytes as an abundant progenitor source for brain macrophage replacement therapies.
Collapse
Affiliation(s)
- Jonathan Bastos
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Carleigh O'Brien
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mónica Vara-Pérez
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Myrthe Mampay
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lynn van Olst
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Liam Barry-Carroll
- School of Biological Sciences, Southampton General Hospital, University of Southampton, Southampton, UK; Nutrineuro, UMR 1286 INRAE, Bordeaux University, Bordeaux INP, Bordeaux, France
| | - Daliya Kancheva
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sophia Leduc
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ayla Line Lievens
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Leen Ali
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Vladislav Vlasov
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Laura Meysman
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hadis Shakeri
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ria Roelandt
- VIB Single Cell Core, VIB, Ghent/Leuven, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Hannah Van Hove
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karen De Vlaminck
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Isabelle Scheyltjens
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Fazeela Yaqoob
- 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; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Maria Breugelmans
- Department of Obstetrics and Prenatal Medicine, UZ Brussel, VUB, Brussels, Belgium
| | - Gilles Faron
- Department of Obstetrics and Prenatal Medicine, UZ Brussel, VUB, Brussels, Belgium
| | - Diego Gomez-Nicola
- School of Biological Sciences, Southampton General Hospital, University of Southampton, Southampton, UK
| | - David Gate
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - F Chris Bennett
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kiavash Movahedi
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
3
|
Chen D, Wang C, Chen X, Li J, Chen S, Li Y, Ma F, Li T, Zou M, Li X, Huang X, Zhang YW, Zhao Y, Bu G, Zheng H, Chen XF, Zhang J, Zhong L. Brain-wide microglia replacement using a nonconditioning strategy ameliorates pathology in mouse models of neurological disorders. Sci Transl Med 2025; 17:eads6111. [PMID: 40305572 DOI: 10.1126/scitranslmed.ads6111] [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: 08/27/2024] [Revised: 01/08/2025] [Accepted: 02/26/2025] [Indexed: 05/02/2025]
Abstract
Growing genetic and pathological evidence has identified microglial dysfunction as a key contributor to the pathogenesis and progression of various neurological disorders, positioning microglia replacement as a promising therapeutic strategy. Traditional bone marrow transplantation (BMT) methods for replenishing brain microglia have limitations, including low efficiency and the potential for brain injury because of preconditioning regimens, such as irradiation or chemotherapy. Moreover, BM-derived cells that migrate to the brain do not recapitulate the phenotypic and functional properties of resident microglia. Here, we present a microglia transplantation strategy devoid of any conditioning, termed "tricyclic microglial depletion for transplantation" (TCMDT). This approach leverages three cycles of microglial depletion using the colony stimulating factor 1 receptor (CSF1R) inhibitor PLX3397, creating an optimal window for efficient engraftment of exogenous microglia. Transplantation of primary cultured microglia by TCMDT successfully restored the identity and functions of endogenous microglia. To evaluate the therapeutic potential of TCMDT, we applied this strategy to two distinct mouse models of neurologic disorder. In a Sandhoff disease model, a neurodegenerative lysosomal storage disorder caused by hexosaminidase subunit beta (Hexb) deficiency, TCMDT effectively replaced deficient microglia, attenuating neurodegeneration and improving motor performance. Similarly, in an Alzheimer's disease (AD)-related amyloid mouse model carrying the triggering receptor expressed on myeloid cells 2 (Trem2) R47H mutation, our transplantation strategy rescued microglial dysfunction and mitigated AD-related pathology. Overall, our study introduces TCMDT as a practical, efficient, and safe approach for microglia replacement, suggesting therapeutic potential for treating neurological disorders associated with microglial dysfunction.
Collapse
Affiliation(s)
- Dadian Chen
- Xiamen Key Laboratory of Brain Center, First Affiliated Hospital of Xiamen University and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Chen Wang
- Department of Neurology and Department of Neuroscience, Xiamen Medical Quality Control Center for Neurology, First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Xi Chen
- Department of Neurosurgery, First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Jiayu Li
- Xiamen Key Laboratory of Brain Center, First Affiliated Hospital of Xiamen University and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Shuai Chen
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Yanzhong Li
- Xiamen Key Laboratory of Brain Center, First Affiliated Hospital of Xiamen University and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Fangling Ma
- Xiamen Key Laboratory of Brain Center, First Affiliated Hospital of Xiamen University and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Tingting Li
- Xiamen Key Laboratory of Brain Center, First Affiliated Hospital of Xiamen University and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Mengling Zou
- Xiamen Key Laboratory of Brain Center, First Affiliated Hospital of Xiamen University and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Xin Li
- Xiamen Key Laboratory of Brain Center, First Affiliated Hospital of Xiamen University and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiaohua Huang
- Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Yun-Wu Zhang
- Xiamen Key Laboratory of Brain Center, First Affiliated Hospital of Xiamen University and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Yingjun Zhao
- Xiamen Key Laboratory of Brain Center, First Affiliated Hospital of Xiamen University and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Guojun Bu
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
| | - Honghua Zheng
- Xiamen Key Laboratory of Brain Center, First Affiliated Hospital of Xiamen University and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiao-Fen Chen
- Xiamen Key Laboratory of Brain Center, First Affiliated Hospital of Xiamen University and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
- Shenzhen Research Institute of Xiamen University, Shenzhen, Guangdong 518063, China
| | - Jie Zhang
- Xiamen Key Laboratory of Brain Center, First Affiliated Hospital of Xiamen University and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Li Zhong
- Xiamen Key Laboratory of Brain Center, First Affiliated Hospital of Xiamen University and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
- Shenzhen Research Institute of Xiamen University, Shenzhen, Guangdong 518063, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, Fujian 361102, China
| |
Collapse
|
4
|
Chung EN, Lee J, Polonio CM, Choi J, Akl CF, Kilian M, Weiß WM, Gunner G, Ye M, Heo TH, Drake SS, Yang L, d'Eca CRGL, Lee JH, Deng L, Farrenkopf D, Schüle AM, Lee HG, Afolabi O, Ghaznavi S, Smirnakis SM, Chiu IM, Kuchroo VK, Quintana FJ, Wheeler MA. Psychedelic control of neuroimmune interactions governing fear. Nature 2025:10.1038/s41586-025-08880-9. [PMID: 40269152 DOI: 10.1038/s41586-025-08880-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 03/11/2025] [Indexed: 04/25/2025]
Abstract
Neuroimmune interactions-signals transmitted between immune and brain cells-regulate many aspects of tissue physiology1, including responses to psychological stress2-5, which can predispose individuals to develop neuropsychiatric diseases6-9. Still, the interactions between haematopoietic and brain-resident cells that influence complex behaviours are poorly understood. Here, we use a combination of genomic and behavioural screens to show that astrocytes in the amygdala limit stress-induced fear behaviour through epidermal growth factor receptor (EGFR). Mechanistically, EGFR expression in amygdala astrocytes inhibits a stress-induced, pro-inflammatory signal-transduction cascade that facilitates neuron-glial crosstalk and stress-induced fear behaviour through the orphan nuclear receptor NR2F2 in amygdala neurons. In turn, decreased EGFR signalling and fear behaviour are associated with the recruitment of meningeal monocytes during chronic stress. This set of neuroimmune interactions is therapeutically targetable through the administration of psychedelic compounds, which reversed the accumulation of monocytes in the brain meninges along with fear behaviour. Together with validation in clinical samples, these data suggest that psychedelics can be used to target neuroimmune interactions relevant to neuropsychiatric disorders and potentially other inflammatory diseases.
Collapse
Affiliation(s)
- Elizabeth N Chung
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Jinsu Lee
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Carolina M Polonio
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Joshua Choi
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Camilo Faust Akl
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Michael Kilian
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Wiebke M Weiß
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Georgia Gunner
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Mingyu Ye
- Department of Neurology, Brigham and Women's Hospital and Jamaica Plain Veterans Administration Hospital, Harvard Medical School, Boston, MA, USA
| | - Tae Hyun Heo
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Sienna S Drake
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Liu Yang
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Catarina R G L d'Eca
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Joon-Hyuk Lee
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Liwen Deng
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Daniel Farrenkopf
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Anton M Schüle
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Hong-Gyun Lee
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Oreoluwa Afolabi
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Sharmin Ghaznavi
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for the Neuroscience of Psychedelics, Massachusetts General Hospital, Boston, MA, USA
| | - Stelios M Smirnakis
- Department of Neurology, Brigham and Women's Hospital and Jamaica Plain Veterans Administration Hospital, Harvard Medical School, Boston, MA, USA
| | - Isaac M Chiu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Vijay K Kuchroo
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Francisco J Quintana
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael A Wheeler
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
5
|
Papetti AV, Jin M, Ma Z, Stillitano AC, Jiang P. Chimeric brain models: Unlocking insights into human neural development, aging, diseases, and cell therapies. Neuron 2025:S0896-6273(25)00256-9. [PMID: 40300597 DOI: 10.1016/j.neuron.2025.03.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/07/2025] [Accepted: 03/31/2025] [Indexed: 05/01/2025]
Abstract
Human-rodent chimeric brain models serve as a unique platform for investigating the pathophysiology of human cells within a living brain environment. These models are established by transplanting human tissue- or human pluripotent stem cell (hPSC)-derived macroglial, microglial, or neuronal lineage cells, as well as cerebral organoids, into the brains of host animals. This approach has opened new avenues for exploring human brain development, disease mechanisms, and regenerative processes. Here, we highlight recent advancements in using chimeric models to study human neural development, aging, and disease. Additionally, we explore the potential applications of these models for studying human glial cell-replacement therapies, studying in vivo human glial-to-neuron reprogramming, and harnessing single-cell omics and advanced functional assays to uncover detailed insights into human neurobiology. Finally, we discuss strategies to enhance the precision and translational relevance of these models, expanding their impact in stem cell and neuroscience research.
Collapse
Affiliation(s)
- Ava V Papetti
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Mengmeng Jin
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Ziyuan Ma
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Alessandro C Stillitano
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Peng Jiang
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA.
| |
Collapse
|
6
|
Goleij P, Amini A, Tabari MAK, Hadipour M, Rezaee A, Daglia M, Aschner M, Sanaye PM, Kumar AP, Khan H. Unraveling the role of the IL-20 cytokine family in neurodegenerative diseases: Mechanisms and therapeutic insights. Int Immunopharmacol 2025; 152:114399. [PMID: 40068518 DOI: 10.1016/j.intimp.2025.114399] [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: 12/10/2024] [Revised: 02/03/2025] [Accepted: 02/27/2025] [Indexed: 03/24/2025]
Abstract
The IL-20 cytokine family, comprising IL-19, IL-20, IL-22, IL-24, and IL-26, has emerged as a critical player in the pathogenesis of neurodegenerative diseases due to its multiple roles in inflammation, tissue repair, and immune modulation. These cytokines signal through IL-20 receptor complexes (IL-20RA/IL-20RB and IL-22RA1/IL-20RB), triggering diverse immune processes. Recent evidence highlights their significant contributions to neuroinflammation and neurodegeneration in central nervous system disorders. IL-20 family cytokines impact microglial activation, which, when dysregulated, exacerbates neuronal damage. Specifically, IL-20 and IL-24 are linked to elevated pro-inflammatory markers in glial cells, promoting neurodegeneration. In contrast, IL-22 exhibits dual functionality, exerting protective and pathological roles depending on the inflammatory milieu. Key mechanisms involve the regulation of blood-brain barrier integrity, oxidative stress, and autophagy. IL-22 and IL-24 also protect neurons by enhancing antioxidant defenses and maintaining epithelial barrier function, while their dysregulation contributes to blood-brain barrier disruption and protein aggregate accumulation, hallmark features of Alzheimer's disease, Parkinson's disease, and multiple sclerosis. Elevated IL-22 levels in Alzheimer's disease and IL-19's regulatory role in multiple sclerosis suggest they may serve as potential biomarkers and therapeutic targets. IL-26's role in amplifying inflammatory cascades further underscores the complexity of this cytokine family in neurodegenerative pathology. Therapeutically, strategies targeting IL-20 cytokines include monoclonal antibodies, receptor modulation, and recombinant cytokine administration. These approaches aim to mitigate neuroinflammation, restore immune balance, and protect neuronal integrity. This review underscores the IL-20 family's emerging relevance in neurodegenerative diseases, highlighting its potential for novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Pouya Goleij
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran; Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Alireza Amini
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Mazandaran 4815733971, Iran
| | - Mohammad Amin Khazeei Tabari
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Mazandaran 4815733971, Iran
| | - Mahboube Hadipour
- Department of Biochemistry, School of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas 7919693116, Iran
| | - Aryan Rezaee
- Medical Doctor, School of Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Maria Daglia
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy; International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China.
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer 209, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| | - Pantea Majma Sanaye
- School of Pharmacy, Zanjan University of Medical Sciences, Zanjan 4513956184, Iran
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore.
| | - Haroon Khan
- Department of Pharmacy, Faculty of Chemical and Life Sciences, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan; Department of Pharmacy, Korea University, Sejong, 20019, South Korea.
| |
Collapse
|
7
|
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
|
8
|
Zhang Y, Du B, Zou M, Peng B, Rao Y. Neuronal Ceroid Lipofuscinosis-Concepts, Classification, and Avenues for Therapy. CNS Neurosci Ther 2025; 31:e70261. [PMID: 39925015 PMCID: PMC11808193 DOI: 10.1111/cns.70261] [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: 10/18/2024] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 02/11/2025] Open
Abstract
Neuronal ceroid lipofuscinosis (NCL) is a group of neurodegenerative lysosomal storage disorders characterized by excessive accumulation of lysosomal lipofuscin. Thirteen subtypes of NCL have been identified, each associated with distinct genes encoding various transmembrane proteins, secretory proteins, or lysosomal enzymes. Clinically, NCL manifests in infants through vision impairment, motor and cognitive dysfunctions, epilepsy, and premature death. The pathological complexity of NCL has hindered the development of effective clinical protocols. Current treatment modalities, including enzyme replacement therapy, pharmacological approaches, gene therapy, and stem cell therapy, have demonstrated limited efficacy. However, emerging evidence suggests a significant relationship between NCL and microglial cells, highlighting the potential of novel microglial cell replacement therapies. This review comprehensively examines the pathogenic genes associated with various NCL subtypes, elucidating their roles, clinical presentations, and corresponding mouse models. Especially, we thoroughly discuss the advances in the clinical study of potential therapeutics, which crucially calls for early diagnosis and treatment more than ever.
Collapse
Affiliation(s)
- Yuheng Zhang
- Department of Neurology, Zhongshan Hospital, Laboratory Animal CenterFudan UniversityShanghaiChina
- Children’s Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory DiseasesFudan UniversityShanghaiChina
| | - Bingying Du
- Children’s Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory DiseasesFudan UniversityShanghaiChina
- Department of NeurologyThe First Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Miaozhan Zou
- Department of Neurology, Zhongshan Hospital, Laboratory Animal CenterFudan UniversityShanghaiChina
- Children’s Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory DiseasesFudan UniversityShanghaiChina
| | - Bo Peng
- Children’s Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory DiseasesFudan UniversityShanghaiChina
| | - Yanxia Rao
- Department of Neurology, Zhongshan Hospital, Laboratory Animal CenterFudan UniversityShanghaiChina
| |
Collapse
|
9
|
Yu Z, Liu J, Liu Z, Liu X, Tuo J, Li J, Tu Y, Tan Q, Ma Y, Bai Y, Xin J, Huang S, Zeng G, Shi A, Wang J, Liu Y, Bu X, Ye L, Wan Y, Liu T, Chen X, Qiu Z, Gao C, Wang Y. Roles of blood monocytes carrying TREM2 R47H mutation in pathogenesis of Alzheimer's disease and its therapeutic potential in APP/PS1 mice. Alzheimers Dement 2025; 21:e14402. [PMID: 39740209 PMCID: PMC11848385 DOI: 10.1002/alz.14402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 09/27/2024] [Accepted: 10/21/2024] [Indexed: 01/02/2025]
Abstract
INTRODUCTION The triggering receptor expressed on myeloid cells 2 (TREM2) arginine-47-histidine (R47H) mutation is a significant risk for Alzheimer's disease (AD) with unclear mechanisms. Previous studies focused on microglial amyloid-β (Aβ) phagocytosis with less attention on the impact of TREM2R47H mutation on blood monocytes. METHODS Bone marrow transplantation (BMT) models were used to assess the contribution of blood monocytes carrying TREM2R47H mutation to AD. RESULTS Aβ phagocytosis was compromised in mouse monocytes carrying the TREM2R47H mutation. Transplantation of bone marrow cells (BMCs) carrying TREM2R47H mutation increased cerebral Aβ burden and aggravated AD-type pathologies. Moreover, the replacement of TREM2R47H-BMCs restored monocytic Aβ phagocytosis, lowered Aβ levels in the blood and brain, and improved cognitive function. DISCUSSION Our study reveals that blood monocytes carrying the TREM2R47H mutation substantially contribute to the pathogenesis of AD, and correcting the TREM2R47H mutation in BMCs would be a potential therapeutic approach for those carrying this mutation. HIGHLIGHTS TREM2R47H mutation compromises the Aβ phagocytosis of blood monocytes. Blood monocytes carrying TREM2R47H mutation contribute substantially to AD pathogenesis. Correction of the TREM2R47H mutation in bone marrow cells ameliorates AD pathologies and cognitive impairments.
Collapse
Affiliation(s)
- Zhong‐Yuan Yu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Institute of Brain and IntelligenceChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Jie Liu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Institute of Brain and IntelligenceChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Zhi‐Hao Liu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xiao‐Yu Liu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Department of NeurologyThe 991st Hospital of Chinese People's Liberation Army Joint Logistic Support ForceXiangyangChina
| | - Jin‐Mei Tuo
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Jiang‐Hui Li
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Yun‐Feng Tu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Qi Tan
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Yuan‐Yuan Ma
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Yu‐Di Bai
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Jia‐Yan Xin
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Shan Huang
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Gui‐Hua Zeng
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - An‐Yu Shi
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Jun Wang
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Yu‐Hui Liu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Xian‐Le Bu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Li‐Lin Ye
- Institute of ImmunologyThird Military Medical UniversityChongqingChina
| | - Ying Wan
- Biomedical Analysis CentreThird Military Medical UniversityChongqingChina
| | - Tong‐Fei Liu
- Institute for Brain Science and DiseaseChongqing Medical UniversityChongqingChina
| | - Xiao‐Wei Chen
- Institute of Brain and IntelligenceChongqingChina
- Brain Research CentreCollaborative Innovation Centre for Brain ScienceThird Military Medical UniversityChongqingChina
| | - Zi‐Long Qiu
- Songjiang HospitalSongjiang InstituteShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Chang‐Yue Gao
- Department of Rehabilitation MedicineDaping Hospital, Third Military Medical UniversityChongqingChina
| | - Yan‐Jiang Wang
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Institute of Brain and IntelligenceChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| |
Collapse
|
10
|
Hu B, Liu X, Xiong S, Gong Q, Yang J, Shi H, Zhang M, Liang F, Zhang Z. Increased cardiac macrophages in Sorbs2-deficient hearts: revealing a potential role for macrophage in responding to embryonic myocardial abnormalities. Front Genet 2025; 15:1525931. [PMID: 39882075 PMCID: PMC11774933 DOI: 10.3389/fgene.2024.1525931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025] Open
Abstract
Macrophages are known to support cardiac development and homeostasis, contributing to tissue remodeling and repair in the adult heart. However, it remains unclear whether embryonic macrophages also respond to abnormalities in the developing heart. Previously, we reported that the structural protein Sorbs2 promotes the development of the second heart field, with its deficiency resulting in atrial septal defects (ASD). In analyzing RNA-seq data, we noted an upregulation of macrophage-related genes in Sorbs2 -/- hearts. Immunostaining and lineage-tracing confirmed an increase in macrophage numbers, underscoring a macrophage response to myocardial abnormalities. Partial depletion of macrophages led to downregulation of genes involved in lipid metabolism, muscle development and organ regeneration, alongside upregulation of genes associated with DNA damage-induced senescence and cardiomyopathy. Additionally, a non-significant increase in septal defects in macrophage-depleted Sorbs2 -/- hearts suggests a potential reparative function for macrophages in maintaining structural integrity. Valve formation, however, remained unaffected. Our findings suggest that embryonic macrophages might sense abnormalities in embryonic cardiomyocytes and could adaptively support cardiac structure and function development in response to myocardial abnormalities.
Collapse
Affiliation(s)
- Beibei Hu
- Pediatric Translational Medicine Institute and Pediatric Congenital Heart Disease Institute, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiangyang Liu
- School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Shanshan Xiong
- Pediatric Translational Medicine Institute and Pediatric Congenital Heart Disease Institute, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qin Gong
- Shanghai United International School (Gubei Campus), Shanghai, China
| | - Junjie Yang
- Pediatric Translational Medicine Institute and Pediatric Congenital Heart Disease Institute, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongjun Shi
- School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Min Zhang
- Pediatric Translational Medicine Institute and Pediatric Congenital Heart Disease Institute, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei Liang
- Neonatal Intensive Care Unit, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Zhang
- Pediatric Translational Medicine Institute and Pediatric Congenital Heart Disease Institute, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Collaborative Innovative Center of Intelligent Medical Device and Active Health, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
11
|
Amann L, Fell A, Monaco G, Sankowski R, Wu HZQ, Jordão MJC, Borst K, Fliegauf M, Masuda T, Ardura-Fabregat A, Paterson N, Nent E, Cook J, Staszewski O, Mossad O, Falk T, Louveau A, Smirnov I, Kipnis J, Lämmermann T, Prinz M. Extrasinusoidal macrophages are a distinct subset of immunologically active dural macrophages. Sci Immunol 2024; 9:eadh1129. [PMID: 39705337 DOI: 10.1126/sciimmunol.adh1129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 11/26/2024] [Indexed: 12/22/2024]
Abstract
Although macrophages in the meningeal compartments of the central nervous system (CNS) have been comprehensively characterized under steady state, studying their contribution to physiological and pathological processes has been hindered by the lack of specific targeting tools in vivo. Recent findings have shown that the dural sinus and its adjacent lymphatic vessels act as a neuroimmune interface. However, the cellular and functional heterogeneity of extrasinusoidal dural macrophages outside this immune hub is not fully understood. Therefore, we comprehensively characterized these cells using single-cell transcriptomics, fate mapping, confocal imaging, clonal analysis, and transgenic mouse lines. Extrasinusoidal dural macrophages were distinct from leptomeningeal and CNS parenchymal macrophages in terms of their origin, expansion kinetics, and transcriptional profiles. During autoimmune neuroinflammation, extrasinusoidal dural macrophages performed efferocytosis of apoptotic granulocytes. Our results highlight a previously unappreciated myeloid cell diversity and provide insights into the brain's innate immune system.
Collapse
Affiliation(s)
- Lukas Amann
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Amelie Fell
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gianni Monaco
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute for Transfusion Medicine and Gene Therapy, Medical Center, University of Freiburg, Freiburg, Germany
| | - Roman Sankowski
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Berta-Ottenstein-Programme for Clinician Scientists, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Huang Zie Quann Wu
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | | | - Katharina Borst
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maximilian Fliegauf
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Takahiro Masuda
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Division of Molecular Neuroimmunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Alberto Ardura-Fabregat
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Neil Paterson
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Elisa Nent
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - James Cook
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ori Staszewski
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Berta-Ottenstein-Programme for Clinician Scientists, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Omar Mossad
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Thorsten Falk
- Department of Computer Sciences, University of Freiburg, Freiburg, Germany
| | - Antoine Louveau
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Igor Smirnov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Brain Immunology and Glia (BIG), Washington University School of Medicine, St. Louis, MO, USA
| | - Jonathan Kipnis
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Brain Immunology and Glia (BIG), Washington University School of Medicine, St. Louis, MO, USA
| | - Tim Lämmermann
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Münster, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| |
Collapse
|
12
|
Shao L, Chang Y, Liu J, Lin L, Chang L, Zhang J, Lan Z, Zhang H, Chen X. scRNA-Seq reveals age-dependent microglial evolution as a determinant of immune response following spinal cord injury. Brain Res Bull 2024; 219:111116. [PMID: 39515654 DOI: 10.1016/j.brainresbull.2024.111116] [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: 08/28/2024] [Revised: 10/24/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Spinal cord injury (SCI) is a debilitating condition of the central nervous system (CNS) that leads to severe impairments in sensory and motor functions. Previous studies have pointed out that patient age is a critical factor influencing SCI prognosis. However, the role of microglia in age-related differences in SCI outcomes remains unclear. The current study aims to identify specific microglial subtypes and investigate their responses and functional differences in SCI recovery across different age groups. Single-cell RNA sequencing (scRNA-seq) data were obtained from the Gene Expression Omnibus (GEO) database, integrating multiple datasets to identify microglial subtypes. We performed pseudotime trajectory analysis and cell-cell communication analysis to understand microglial differentiation and interactions. Finally, immunofluorescence staining of mouse model samples was conducted to validate our bioinformatics findings. Microglia were classified into four subtypes: Homeostatic, Proliferating, Inflammatory A, and Inflammatory B. The Young SCI group exhibited a higher proportion of Homeostatic microglia and Inflammatory microglia A, whereas the old SCI group had more Inflammatory Microglia B but lacked Homeostatic Microglia. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses revealed that markers for homeostasis microglia were enriched in immune modulation pathways. While makers for Inflammatory Microglia were enriched in immune response pathways. Specifically, markers for Inflammatory microglia B were enriched in pathways associated with overactive immune response. Pseudotime analysis indicated that microglia in young mice predominantly differentiated into Inflammatory Microglia A and Homeostatic Microglia, whereas in old mice, they tended to only differentiate into Inflammatory Microglia B. CellChat analysis showed increased pro-inflammatory signaling generated by Inflammatory Microglia B, exclusively in the old group. Our study demonstrates significant differences in microglial subtypes and functions between different age groups following SCI. These findings provide novel insights into the development of age-related therapeutic strategies and microglia-targeted biological treatments for SCI.
Collapse
Affiliation(s)
- Lufei Shao
- Neurology Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China; Ningxia nervous system disease Diagnosis and treatment Engineering Technology Research center, Yinchuan 750004, China
| | - Yueliang Chang
- Neurology Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Jinfang Liu
- Neurology Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Leilei Lin
- Orthopedics Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Long Chang
- Orthopedics Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Jialin Zhang
- Orthopedics Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Zhibin Lan
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Honglai Zhang
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Xiaolei Chen
- Orthopedics Department, General Hospital of Ningxia Medical University, Yinchuan 750004, China; Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
13
|
Montepeloso A, Mattioli D, Pellin D, Peviani M, Genovese P, Biffi A. Haploinsufficiency at the CX3CR1 locus of hematopoietic stem cells favors the appearance of microglia-like cells in the central nervous system of transplant recipients. Nat Commun 2024; 15:10192. [PMID: 39587072 PMCID: PMC11589136 DOI: 10.1038/s41467-024-54515-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 11/13/2024] [Indexed: 11/27/2024] Open
Abstract
Transplantation of engineered hematopoietic stem/progenitor cells (HSPCs) showed curative potential in patients affected by neurometabolic diseases treated in early stage. Favoring the engraftment and maturation of the engineered HSPCs in the central nervous system (CNS) could allow enhancing further the therapeutic potential of this approach. Here we unveil that HSPCs haplo-insufficient at the Cx3cr1 (Cx3cr1-/+) locus are favored in central nervous system (CNS) engraftment and generation of microglia-like progeny cells (MLCs) as compared to wild type (Cx3cr1+/+) HSPCs upon transplantation in mice. Based on this evidence, we have developed a CRISPR-based targeted gene addition strategy at the human CX3CR1 locus resulting in an enhanced ability of the edited human HSPCs to generate mature MLCs upon transplantation in immunodeficient mice, and in lineage specific, regulated and robust transgene expression. This approach, which benefits from the modulation of pathways involved in microglia maturation and migration in haplo-insufficient cells, may broaden the application of HSPC gene therapy to a larger spectrum of neurometabolic and neurodegenerative diseases.
Collapse
Affiliation(s)
- Annita Montepeloso
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
- Gene Therapy Consulting, Padua, Italy
| | - Davide Mattioli
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Padua, Padua, Italy
| | - Danilo Pellin
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Marco Peviani
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Pietro Genovese
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Alessandra Biffi
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Padua, Padua, Italy.
| |
Collapse
|
14
|
Michalides BA, Shoger KE, Kruszelnicki S, Cheemalavagu N, Martinez-Turak A, Jackson-Strong M, Laughlin CR, Betsur OS, Colby D, Meisel M, Gingras S, Gottschalk RA. Fth1-mScarlet Reports Monocyte State during Lipopolysaccharide-induced Lung Inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1508-1515. [PMID: 39392393 PMCID: PMC11534542 DOI: 10.4049/jimmunol.2400215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/14/2024] [Indexed: 10/12/2024]
Abstract
Monocytes and macrophages are central to host defense but also contribute to inflammation-associated pathology. Efforts to manipulate monocyte and macrophage function are limited by our ability to effectively quantify the functional programs of these cells. We identified the gene Fth1, which encodes the ferritin H chain, as highly predictive of alveolar macrophage transcriptomic states during LPS-induced lung inflammation and developed an Fth1-mScarlet reporter mouse. In the steady-state lung, high Fth1-mScarlet expression is restricted to alveolar macrophages. In response to LPS-induced lung inflammation, Fth1 reporter activity is robustly increased in monocytes, with its expression reporting genes that are differentially expressed in monocytes versus macrophages. Consistent with this reporter-associated gene profile, within the Lyz2-GFP+CD11b+Ly6C+ gate, the highest Fth1 reporter expression was observed in CD11c+ cells, indicative of monocyte-to-macrophage differentiation. Although Fth1-mScarlet was induced in monocytes responding to either TLR4 ligation or M-CSF-induced macrophage differentiation in vitro, TLR4-dependent expression occurred with greater speed and magnitude. Considering this, we suggest that Fth1-mScarlet expression reports monocyte-to-macrophage differentiation, with increased expression in proinflammatory states. Dissecting macrophage differentiation from inflammatory programs will be enhanced when combining Fth1-mScarlet with other reporter systems. Thus, the Fth1-mScarlet model addresses an important lack of tools to report the diverse spectrum of monocyte and macrophage states in vivo.
Collapse
Affiliation(s)
- Brandon A. Michalides
- University of Pittsburgh, Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA USA
| | - Karsen E. Shoger
- University of Pittsburgh, Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA USA
| | - Sonia Kruszelnicki
- University of Pittsburgh, Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA USA
| | - Neha Cheemalavagu
- University of Pittsburgh, Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA USA
| | - Anamarie Martinez-Turak
- University of Pittsburgh, Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA USA
| | - Morgan Jackson-Strong
- University of Pittsburgh, Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA USA
| | - Colin R. Laughlin
- University of Pittsburgh, Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA USA
| | - Omkar S. Betsur
- University of Pittsburgh, Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA USA
| | - Devon Colby
- University of Pittsburgh, Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA USA
| | - Marlies Meisel
- University of Pittsburgh, Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA USA
| | - Sebastien Gingras
- University of Pittsburgh, Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA USA
| | - Rachel A. Gottschalk
- University of Pittsburgh, Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA USA
| |
Collapse
|
15
|
Zhou H, Yang RK, Li Q, Li Z, Wang YC, Li SY, Miao Y, Sun XH, Wang Z. MicroRNA-146a-5p protects retinal ganglion cells through reducing neuroinflammation in experimental glaucoma. Glia 2024; 72:2115-2141. [PMID: 39041109 DOI: 10.1002/glia.24600] [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: 01/30/2024] [Revised: 06/27/2024] [Accepted: 07/15/2024] [Indexed: 07/24/2024]
Abstract
Neuroinflammation plays important roles in retinal ganglion cell (RGC) degeneration in glaucoma. MicroRNA-146 (miR-146) has been shown to regulate inflammatory response in neurodegenerative diseases. In this study, whether and how miR-146 could affect RGC injury in chronic ocular hypertension (COH) experimental glaucoma were investigated. We showed that in the members of miR-146 family only miR-146a-5p expression was upregulated in COH retinas. The upregulation of miR-146a-5p was observed in the activated microglia and Müller cells both in primary cultured conditions and in COH retinas, but mainly occurred in microglia. Overexpression of miR-146a-5p in COH retinas reduced the levels pro-inflammatory cytokines and upregulated the levels of anti-inflammatory cytokines, which were further confirmed in the activated primary cultured microglia. Transfection of miR-146a-5p mimic increased the percentage of anti-inflammatory phenotype in the activated BV2 microglia, while transfection of miR-146a-5p inhibitor resulted in the opposite effects. Transfection of miR-146a-5p mimic/agomir inhibited the levels of interleukin-1 receptor associated kinase (IRAK1) and TNF receptor associated factor 6 (TRAF6) and phosphorylated NF-κB subunit p65. Dual luciferase reporter gene assay confirmed that miR-146a-5p could directly target IRAK1 and TRAF6. Moreover, downregulation of IRAK1 and TRAF6 by siRNA techniques or blocking NF-κB by SN50 in cultured microglia reversed the miR-146a-5p inhibitor-induced changes of inflammatory cytokines. In COH retinas, overexpression of miR-146a-5p reduced RGC apoptosis, increased RGC survival, and partially rescued the amplitudes of photopic negative response. Our results demonstrate that overexpression of miR-146a-5p attenuates RGC injury in glaucoma by reducing neuroinflammation through downregulating IRAK1/TRAF6/NF-κB signaling pathway in microglia.
Collapse
Affiliation(s)
- Han Zhou
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Rui-Kang Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Qian Li
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, NHC Key Laboratory of Myopia, Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Zhen Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yong-Chen Wang
- Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Shu-Ying Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yanying Miao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xing-Huai Sun
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, NHC Key Laboratory of Myopia, Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Zhongfeng Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Wang M, Dufort C, Du Z, Shi R, Xu F, Huang Z, Sigler AR, Leak RK, Hu X. IL-33/ST2 signaling in monocyte-derived macrophages maintains blood-brain barrier integrity and restricts infarctions early after ischemic stroke. J Neuroinflammation 2024; 21:274. [PMID: 39449077 PMCID: PMC11515348 DOI: 10.1186/s12974-024-03264-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Brain microglia and infiltrating monocyte-derived macrophages are vital in preserving blood vessel integrity after stroke. Understanding mechanisms that induce immune cells to adopt vascular-protective phenotypes may hasten the development of stroke treatments. IL-33 is a potent chemokine released from damaged cells, such as CNS glia after stroke. The activation of IL-33/ST2 signaling has been shown to promote neuronal viability and white matter integrity after ischemic stroke. The impact of IL-33/ST2 on blood-brain barrier (BBB) integrity, however, remains unknown. The current study fills this gap and reveals a critical role of IL-33/ST2 signaling in macrophage-mediated BBB protection after stroke. METHODS Transient middle cerebral artery occlusion (tMCAO) was performed to induce ischemic stroke in wildtype (WT) versus ST2 knockout (KO) male mice. IL-33 was applied intranasally to tMCAO mice with or without dietary PLX5622 to deplete microglia/macrophages. ST2 KO versus WT bone marrow or macrophage cell transplantations were used to test the involvement of ST2+ macrophages in BBB integrity. Macrophages were cocultured in transwells with brain endothelial cells (ECs) after oxygen-glucose deprivation (OGD) to test potential direct effects of IL33-treated macrophages on the BBB in vitro. RESULTS The ST2 receptor was expressed in brain ECs, microglia, and infiltrating macrophages. Global KO of ST2 led to more IgG extravasation and loss of ZO-1 in cerebral microvessels 3 days post-tMCAO. Intranasal IL-33 administration reduced BBB leakage and infarct severity in microglia/macrophage competent mice, but not in microglia/macrophage depleted mice. Worse BBB injury was observed after tMCAO in chimeric WT mice reconstituted with ST2 KO bone marrow, and in WT mice whose monocytes were replaced by ST2 KO monocytes. Macrophages treated with IL-33 reduced in vitro barrier leakage and maintained tight junction integrity after OGD. In contrast, IL-33 exerted minimal direct effects on the endothelial barrier in the absence of macrophages. IL-33-treated macrophages demonstrated transcriptional upregulation of an array of protective factors, suggesting a shift towards favorable phenotypes. CONCLUSION Our results demonstrate that early-stage IL-33/ST2 signaling in infiltrating macrophages reduces the extent of acute BBB disruption after stroke. Intranasal IL-33 administration may represent a new strategy to reduce BBB leakage and infarct severity.
Collapse
Affiliation(s)
- Miao Wang
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, 200 Lothrop Street, SBST 506, Pittsburgh, PA, 15213, USA
| | - Connor Dufort
- Department of Neurology, School of Medicine, University of Pittsburgh, 200 Lothrop Street, SBST 506, Pittsburgh, PA, 15213, USA
| | - Zhihong Du
- Department of Neurology, School of Medicine, University of Pittsburgh, 200 Lothrop Street, SBST 506, Pittsburgh, PA, 15213, USA
| | - Ruyu Shi
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Fei Xu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, 200 Lothrop Street, SBST 506, Pittsburgh, PA, 15213, USA
| | - Zhentai Huang
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, 200 Lothrop Street, SBST 506, Pittsburgh, PA, 15213, USA
| | - Ana Rios Sigler
- Department of Neurology, School of Medicine, University of Pittsburgh, 200 Lothrop Street, SBST 506, Pittsburgh, PA, 15213, USA
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, 15282, USA
| | - Xiaoming Hu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA.
- Department of Neurology, School of Medicine, University of Pittsburgh, 200 Lothrop Street, SBST 506, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
17
|
Cao J, Chen H, Zhang Y, Kang Y, Zhou S, Liao Z, Gao L, Yin J, Jing Y. Androgen deprivation exacerbates AD pathology by promoting the loss of microglia in an age-dependent manner. Life Sci 2024; 355:122973. [PMID: 39142510 DOI: 10.1016/j.lfs.2024.122973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/25/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024]
Abstract
AIMS Microglial cells are integral to the pathogenesis of Alzheimer's disease (AD). The observed sex disparity in AD prevalence, with a notable predominance in women, implies a potential influence of sex hormones, such as androgens, on disease mechanisms. Despite this, the specific effects of androgens on microglia remain unclear. This study is designed to delineate the interplay between androgens and the survival and inflammatory profile of microglial cells, as well as to explore their contribution to the progression of AD. METHODS AND KEY FINDINGS To create a chronic androgen deficiency model, 3-month-old wild-type (WT) mice and APP/PS1 mice underwent bilateral orchiectomy (ORX), with age-matched sham-operated controls. Cognitive and memory were evaluated at 5 and 12 months, paralleled by assessments of amyloid-beta (Aβ) and microglial morphology in hippocampal and cortical areas. The ORX treatment in mice resulted in diminished microglial populations and morphological alterations, alongside an increase in Aβ plaques and a concomitant decline in cognitive performance that exacerbated over time. In vitro, dihydrotestosterone (DHT) was found to stimulate microglial proliferation and ameliorate Aβ1-42-induced apoptosis. SIGNIFICANCE These findings suggested that androgens may exert a protective role, maintaining the normal proliferation and functionality of microglial cells. This preservation could potentially slow the progression of AD. As a result, our study provided a conceptual framework for the development of novel therapeutic strategies for AD.
Collapse
Affiliation(s)
- Jiaxin Cao
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Haichao Chen
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Yishu Zhang
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Yiting Kang
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Siwei Zhou
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Zirui Liao
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Liping Gao
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Jie Yin
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Yuhong Jing
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China; Key Laboratory of Preclinical Study for New Drugs of Gansu province, Lanzhou University, Lanzhou, Gansu, People's Republic of China.
| |
Collapse
|
18
|
Schiel V, Bhattacharya R, Gupta A, Eftekharian K, Xia A, Santa Maria PL. Targeting the NLRP3 inflammasome in cochlear macrophages protects against hearing loss in chronic suppurative otitis media. J Neuroinflammation 2024; 21:223. [PMID: 39277762 PMCID: PMC11402200 DOI: 10.1186/s12974-024-03212-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/09/2024] [Indexed: 09/17/2024] Open
Abstract
The activation of the NLRP3 inflammasome has been linked to several inflammatory and autoinflammatory diseases. Despite cases of potential hearing improvement in immune-mediated diseases, direct evidence of the efficacy of targeting this mechanism in the inner ear is still lacking. Previously, we discovered that macrophages are associated with Sensorineural Hearing loss (SNHL) in Chronic Suppurative Otitis Media (CSOM), the leading cause of this permanent hearing loss in the developing world and incurring costs of $4 to $11 billion dollars in the United States. However, the underlying mechanism remained unknown. Here, we investigate how macrophages drive permanent hearing loss in CSOM. We first confirmed the occurrence of NLRP3 inflammasome activation in cochlear macrophages in CSOM. We then revealed that Outer Hair Cells (OHCs) were protected in CSOM by macrophage depletion and subsequently confirmed the same protection in the NLRP3 knockout condition. Furthermore, we showed that therapeutic inhibition of NLRP3 inflammasome activation and downstream inhibition of IL-1β protects OHCs in CSOM. Collectively, our data demonstrates that the main driver for hearing loss in CSOM is NLRP3 inflammasome activation in cochlear macrophages and this is therapeutically targetable, leading the way for the development of interventions to prevent the leading cause of permanent hearing loss and a costly disease in the developed world.
Collapse
Affiliation(s)
- Viktoria Schiel
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University, Palo Alto, CA, 94305, USA
| | - Ritwija Bhattacharya
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University, Palo Alto, CA, 94305, USA
| | - Ankur Gupta
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University, Palo Alto, CA, 94305, USA
| | - Kourosh Eftekharian
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University, Palo Alto, CA, 94305, USA
| | - Anping Xia
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University, Palo Alto, CA, 94305, USA.
| | - Peter L Santa Maria
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University, Palo Alto, CA, 94305, USA.
| |
Collapse
|
19
|
Milazzo R, Montepeloso A, Kumar R, Ferro F, Cavalca E, Rigoni P, Cabras P, Ciervo Y, Das S, Capotondo A, Pellin D, Peviani M, Biffi A. Therapeutic efficacy of intracerebral hematopoietic stem cell gene therapy in an Alzheimer's disease mouse model. Nat Commun 2024; 15:8024. [PMID: 39271711 PMCID: PMC11399302 DOI: 10.1038/s41467-024-52301-w] [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: 10/03/2022] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
The conditions supporting the generation of microglia-like cells in the central nervous system (CNS) after transplantation of hematopoietic stem/progenitor cells (HSPC) have been studied to advance the treatment of neurodegenerative disorders. Here, we explored the transplantation efficacy of different cell subsets and delivery routes with the goal of favoring the establishment of a stable and exclusive engraftment of HSPCs and their progeny in the CNS of female mice. In this setting, we show that the CNS environment drives the expansion, distribution and myeloid differentiation of the locally transplanted cells towards a microglia-like phenotype. Intra-CNS transplantation of HSPCs engineered to overexpress TREM2 decreased neuroinflammation, Aβ aggregation and improved memory in 5xFAD female mice. Our proof of concept study demonstrates the therapeutic potential of HSPC gene therapy for Alzheimer's disease.
Collapse
Affiliation(s)
- Rita Milazzo
- Division of Hematology, Oncology and Stem Cell Transplantation, Department of Women and Child's Health, University of Padua, Padua, Italy
- San Raffaele Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cell and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Annita Montepeloso
- Gene Therapy Program, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Rajesh Kumar
- Gene Therapy Program, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Francesca Ferro
- San Raffaele Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cell and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
- Gene Therapy Program, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Eleonora Cavalca
- San Raffaele Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cell and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
- Gene Therapy Program, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Pietro Rigoni
- Division of Hematology, Oncology and Stem Cell Transplantation, Department of Women and Child's Health, University of Padua, Padua, Italy
| | - Paolo Cabras
- Department of Biology and Biotechnology "L. Spallanzani", Cellular and Molecular Neuropharmacology lab, University of Pavia, Pavia, Italy
| | - Yuri Ciervo
- Division of Hematology, Oncology and Stem Cell Transplantation, Department of Women and Child's Health, University of Padua, Padua, Italy
| | - Sabyasachi Das
- Gene Therapy Program, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Alessia Capotondo
- San Raffaele Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cell and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Danilo Pellin
- Gene Therapy Program, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Marco Peviani
- Gene Therapy Program, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- Department of Biology and Biotechnology "L. Spallanzani", Cellular and Molecular Neuropharmacology lab, University of Pavia, Pavia, Italy
| | - Alessandra Biffi
- Division of Hematology, Oncology and Stem Cell Transplantation, Department of Women and Child's Health, University of Padua, Padua, Italy.
- San Raffaele Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cell and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy.
- Gene Therapy Program, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA.
| |
Collapse
|
20
|
Rao Y, Peng B. Microglia bridge brain activity and blood pressure. Immunity 2024; 57:2000-2002. [PMID: 39260350 DOI: 10.1016/j.immuni.2024.08.006] [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: 07/31/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/13/2024]
Abstract
Our brain is not an immune-privileged island isolated from peripheries, but how non-neuronal brain cells interact with the peripheral system is not well understood. Wei et al. report that microglia in the hypothalamic paraventricular nucleus (PVN) with unique vasculature can detect ATP derived from hemodynamic disturbance. These microglia in the PVN regulate the response to hypertension via ATP-P2Y12-C/EBPβ signaling.
Collapse
Affiliation(s)
- Yanxia Rao
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, Department of Laboratory Animal Science, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200040, China
| | - Bo Peng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, Department of Laboratory Animal Science, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200040, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China.
| |
Collapse
|
21
|
Piccioni G, Maisto N, d'Ettorre A, Strimpakos G, Nisticò R, Triaca V, Mango D. Switch to phagocytic microglia by CSFR1 inhibition drives amyloid-beta clearance from glutamatergic terminals rescuing LTP in acute hippocampal slices. Transl Psychiatry 2024; 14:338. [PMID: 39179543 PMCID: PMC11344079 DOI: 10.1038/s41398-024-03019-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 06/21/2024] [Accepted: 07/08/2024] [Indexed: 08/26/2024] Open
Abstract
Microglia, traditionally regarded as innate immune cells in the brain, drive neuroinflammation and synaptic dysfunctions in the early phases of Alzheimer disease (AD), acting upstream to Aβ accumulation. Colony stimulating factor 1-receptor (CSF-1R) is predominantly expressed on microglia and its levels are significantly increased in neurodegenerative diseases, possibly contributing to the chronic inflammatory microglial response. On the other hand, CSF-1R inhibitors confer neuroprotection in preclinical models of neurodegenerative diseases. Here, we determined the effects of the CSF-1R inhibitor PLX3397 on the Aβ-mediated synaptic alterations in ex vivo hippocampal slices. Electrophysiological findings show that PLX3397 rescues LTP impairment and neurotransmission changes induced by Aβ. In addition, using confocal imaging experiments, we demonstrate that PLX3397 stimulates a microglial transition toward a phagocytic phenotype, which in turn promotes the clearance of Aβ from glutamatergic terminals. We believe that the selective pruning of Aβ-loaded synaptic terminals might contribute to the restoration of LTP and excitatory transmission alterations observed upon acute PLX3397 treatment. This result is in accordance with the mechanism proposed for CSF1R inhibitors, that is to eliminate responsive microglia and replace it with newly generated, homeostatic microglia, capable of promoting brain repair. Overall, our findings identify a connection between the rapid microglia adjustments and the early synaptic alterations observed in AD, possibly highlighting a novel disease-modifying target.
Collapse
Affiliation(s)
- Gaia Piccioni
- Laboratory Pharmacology of Synaptic Plasticity, European Brain Research Institute, Rome, Italy
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Nunzia Maisto
- Laboratory Pharmacology of Synaptic Plasticity, European Brain Research Institute, Rome, Italy
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Asia d'Ettorre
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), International Campus A. Buzzati-Traverso, Rome, Italy
- School of Pharmacy, University of Rome "Tor Vergata", Rome, Italy
| | - Georgios Strimpakos
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), International Campus A. Buzzati-Traverso, Rome, Italy
| | - Robert Nisticò
- Laboratory Pharmacology of Synaptic Plasticity, European Brain Research Institute, Rome, Italy.
- School of Pharmacy, University of Rome "Tor Vergata", Rome, Italy.
| | - Viviana Triaca
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), International Campus A. Buzzati-Traverso, Rome, Italy.
| | - Dalila Mango
- Laboratory Pharmacology of Synaptic Plasticity, European Brain Research Institute, Rome, Italy.
- School of Pharmacy, University of Rome "Tor Vergata", Rome, Italy.
| |
Collapse
|
22
|
Becker SH, Ronayne CE, Bold TD, Jenkins MK. CD4 + T cells recruit, then engage macrophages in cognate interactions to clear Mycobacterium tuberculosis from the lungs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.22.609198. [PMID: 39229103 PMCID: PMC11370583 DOI: 10.1101/2024.08.22.609198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
IFN-γ-producing CD4 + T cells are required for protection against lethal Mycobacterium tuberculosis ( Mtb ) infections. However, the ability of CD4 + T cells to suppress Mtb growth cannot be fully explained by IFN-γ or other known T cell products. In this study, we show that CD4 + T cell-derived IFN-γ promoted the recruitment of monocyte-derived macrophages (MDMs) to the lungs of Mtb -infected mice. Although the recruited MDMs became quickly and preferentially infected with Mtb , CD4 + T cells rapidly disinfected the MDMs. Clearance of Mtb from MDMs was not explained by IFN-γ, but rather by MHCII-mediated cognate interactions with CD4 + T cells. These interactions promoted MDM expression of glycolysis genes essential for Mtb control. Thus, by recruiting MDMs, CD4 + T cells initiate a cycle of bacterial phagocytosis, Mtb antigen presentation and disinfection in an attempt to clear the bacteria from the lungs.
Collapse
|
23
|
Gupta AO, Furcich JW, Nascene DR, Kemp S, King CJ, Nolan EE, Durose W, Miller BS, Orchard PJ, Lund TC. Targeting VEGF-mediated blood-brain barrier disruption in advanced cerebral leukodystrophy. J Neuroimmunol 2024; 393:578395. [PMID: 38897089 DOI: 10.1016/j.jneuroim.2024.578395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/13/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024]
Abstract
The earliest clinical manifestation of cerebral adrenoleukodystrophy (CALD) is adrenal insufficiency (AI) characterized by elevations in ACTH and loss of cortisol. We showed high (though physiologically achievable) levels of ACTH increases endothelial permeability, increases anisotropy, and increases VEGF secretion. An ACBD1 knockout endothelial cell line had increased sensitivity to ACTH and VEGF. Inhibition of VEGF via application of anti-VEGF (bevacizumab) improved permeability. Six boys with advanced CALD were treated with bevacizumab combined with dexamethasone and ruxolitinib as immune suppressants. Most boys had decreases in gadolinium enhancement on MRI indicating improvement in endothelial function, though all boys continued to progress symptomatically.
Collapse
Affiliation(s)
- Ashish O Gupta
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America
| | - Justin W Furcich
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America
| | - David R Nascene
- Department of Diagnostic Radiology, University of Minnesota Medical Center, Minneapolis, MN 55455, United States of America
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, the Netherlands
| | - Carina J King
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America
| | - Erin E Nolan
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America
| | - Willa Durose
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America
| | - Bradley S Miller
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America
| | - Paul J Orchard
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America
| | - Troy C Lund
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America.
| |
Collapse
|
24
|
Du S, Drieu A, Cheng Y, Storck SE, Rustenhoven J, Mamuladze T, Bhattarai B, Brioschi S, Nguyen K, Ou F, Cao J, Rodrigues PF, Smirnov I, DeNardo D, Ginhoux F, Cella M, Colonna M, Kipnis J. Brain-Engrafted Monocyte-derived Macrophages from Blood and Skull-Bone Marrow Exhibit Distinct Identities from Microglia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.08.606900. [PMID: 39211090 PMCID: PMC11361186 DOI: 10.1101/2024.08.08.606900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia are thought to originate exclusively from primitive macrophage progenitors in the yolk sac (YS) and to persist throughout life without much contribution from definitive hematopoiesis. Here, using lineage tracing, pharmacological manipulation, and RNA-sequencing, we elucidated the presence and characteristics of monocyte-derived macrophages (MDMs) in the brain parenchyma at baseline and during microglia repopulation, and defined the core transcriptional signatures of brain-engrafted MDMs. Lineage tracing mouse models revealed that MDMs transiently express CD206 during brain engraftment as CD206 + microglia precursors in the YS. We found that brain-engrafted MDMs exhibit transcriptional and epigenetic characteristics akin to meningeal macrophages, likely due to environmental imprinting within the meningeal space. Utilizing parabiosis and skull transplantation, we demonstrated that monocytes from both peripheral blood and skull bone marrow can repopulate microglia-depleted brains. Our results reveal the heterogeneous origins and cellular dynamics of brain parenchymal macrophages at baseline and in models of microglia depletion.
Collapse
|
25
|
Liu Z, Liu M, Xiong Y, Wang Y, Bu X. Crosstalk between bone and brain in Alzheimer's disease: Mechanisms, applications, and perspectives. Alzheimers Dement 2024; 20:5720-5739. [PMID: 38824621 PMCID: PMC11350061 DOI: 10.1002/alz.13864] [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: 02/05/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 06/04/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that involves multiple systems in the body. Numerous recent studies have revealed bidirectional crosstalk between the brain and bone, but the interaction between bone and brain in AD remains unclear. In this review, we summarize human studies of the association between bone and brain and provide an overview of their interactions and the underlying mechanisms in AD. We review the effects of AD on bone from the aspects of AD pathogenic proteins, AD risk genes, neurohormones, neuropeptides, neurotransmitters, brain-derived extracellular vesicles (EVs), and the autonomic nervous system. Correspondingly, we elucidate the underlying mechanisms of the involvement of bone in the pathogenesis of AD, including bone-derived hormones, bone marrow-derived cells, bone-derived EVs, and inflammation. On the basis of the crosstalk between bone and the brain, we propose potential strategies for the management of AD with the hope of offering novel perspectives on its prevention and treatment. HIGHLIGHTS: The pathogenesis of AD, along with its consequent changes in the brain, may involve disturbing bone homeostasis. Degenerative bone disorders may influence the progression of AD through a series of pathophysiological mechanisms. Therefore, relevant bone intervention strategies may be beneficial for the comprehensive management of AD.
Collapse
Affiliation(s)
- Zhuo‐Ting Liu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease (Third Military Medical University)ChongqingChina
| | - Ming‐Han Liu
- Department of OrthopaedicsXinqiao Hospital, Third Military Medical UniversityChongqingChina
| | - Yan Xiong
- Department of OrthopaedicsDaping Hospital, Third Military Medical UniversityChongqingChina
| | - Yan‐Jiang Wang
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease (Third Military Medical University)ChongqingChina
- Institute of Brain and IntelligenceThird Military Medical UniversityChongqingChina
| | - Xian‐Le Bu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease (Third Military Medical University)ChongqingChina
- Institute of Brain and IntelligenceThird Military Medical UniversityChongqingChina
| |
Collapse
|
26
|
Hu A, Schmidt MHH, Heinig N. Microglia in retinal angiogenesis and diabetic retinopathy. Angiogenesis 2024; 27:311-331. [PMID: 38564108 PMCID: PMC11303477 DOI: 10.1007/s10456-024-09911-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 02/18/2024] [Indexed: 04/04/2024]
Abstract
Diabetic retinopathy has a high probability of causing visual impairment or blindness throughout the disease progression and is characterized by the growth of new blood vessels in the retina at an advanced, proliferative stage. Microglia are a resident immune population in the central nervous system, known to play a crucial role in regulating retinal angiogenesis in both physiological and pathological conditions, including diabetic retinopathy. Physiologically, they are located close to blood vessels and are essential for forming new blood vessels (neovascularization). In diabetic retinopathy, microglia become widely activated, showing a distinct polarization phenotype that leads to their accumulation around neovascular tufts. These activated microglia induce pathogenic angiogenesis through the secretion of various angiogenic factors and by regulating the status of endothelial cells. Interestingly, some subtypes of microglia simultaneously promote the regression of neovascularization tufts and normal angiogenesis in neovascularization lesions. Modulating the state of microglial activation to ameliorate neovascularization thus appears as a promising potential therapeutic approach for managing diabetic retinopathy.
Collapse
Affiliation(s)
- Aiyan Hu
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Fetscherstr 74, 01307, Dresden, Germany
| | - Mirko H H Schmidt
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Fetscherstr 74, 01307, Dresden, Germany.
| | - Nora Heinig
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Fetscherstr 74, 01307, Dresden, Germany.
| |
Collapse
|
27
|
Colella P, Sayana R, Suarez-Nieto MV, Sarno J, Nyame K, Xiong J, Pimentel Vera LN, Arozqueta Basurto J, Corbo M, Limaye A, Davis KL, Abu-Remaileh M, Gomez-Ospina N. CNS-wide repopulation by hematopoietic-derived microglia-like cells corrects progranulin deficiency in mice. Nat Commun 2024; 15:5654. [PMID: 38969669 PMCID: PMC11226701 DOI: 10.1038/s41467-024-49908-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 06/17/2024] [Indexed: 07/07/2024] Open
Abstract
Hematopoietic stem cell transplantation can deliver therapeutic proteins to the central nervous system (CNS) through transplant-derived microglia-like cells. However, current conditioning approaches result in low and slow engraftment of transplanted cells in the CNS. Here we optimized a brain conditioning regimen that leads to rapid, robust, and persistent microglia replacement without adverse effects on neurobehavior or hematopoiesis. This regimen combines busulfan myeloablation and six days of Colony-stimulating factor 1 receptor inhibitor PLX3397. Single-cell analyses revealed unappreciated heterogeneity of microglia-like cells with most cells expressing genes characteristic of homeostatic microglia, brain-border-associated macrophages, and unique markers. Cytokine analysis in the CNS showed transient inductions of myeloproliferative and chemoattractant cytokines that help repopulate the microglia niche. Bone marrow transplant of progranulin-deficient mice conditioned with busulfan and PLX3397 restored progranulin in the brain and eyes and normalized brain lipofuscin storage, proteostasis, and lipid metabolism. This study advances our understanding of CNS repopulation by hematopoietic-derived cells and demonstrates its therapeutic potential for treating progranulin-dependent neurodegeneration.
Collapse
Affiliation(s)
- Pasqualina Colella
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Ruhi Sayana
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | | | - Jolanda Sarno
- Hematology, Oncology, Stem Cell Transplant, and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900, Monza, Italy
| | - Kwamina Nyame
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA, 94305, USA
| | - Jian Xiong
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA, 94305, USA
| | | | | | - Marco Corbo
- MedGenome, Inc, 348 Hatch Dr, Foster City, CA, 94404, USA
| | - Anay Limaye
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
- MedGenome, Inc, 348 Hatch Dr, Foster City, CA, 94404, USA
| | - Kara L Davis
- Hematology, Oncology, Stem Cell Transplant, and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA
| | - Monther Abu-Remaileh
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA, 94305, USA
| | - Natalia Gomez-Ospina
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
28
|
Liu Y, Xia X, Zheng M, Shi B. Bio-Nano Toolbox for Precision Alzheimer's Disease Gene Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2314354. [PMID: 38778446 DOI: 10.1002/adma.202314354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 05/01/2024] [Indexed: 05/25/2024]
Abstract
Alzheimer's disease (AD) is the most burdensome aging-associated neurodegenerative disorder, and its treatment encounters numerous failures during drug development. Although there are newly approved in-market β-amyloid targeting antibody solutions, pathological heterogeneity among patient populations still challenges the treatment outcome. Emerging advances in gene therapies offer opportunities for more precise personalized medicine; while, major obstacles including the pathological heterogeneity among patient populations, the puzzled mechanism for druggable target development, and the precision delivery of functional therapeutic elements across the blood-brain barrier remain and limit the use of gene therapy for central neuronal diseases. Aiming for "precision delivery" challenges, nanomedicine provides versatile platforms that may overcome the targeted delivery challenges for AD gene therapy. In this perspective, to picture a toolbox for AD gene therapy strategy development, the most recent advances from benchtop to clinics are highlighted, possibly available gene therapy targets, tools, and delivery platforms are outlined, their challenges as well as rational design elements are addressed, and perspectives in this promising research field are discussed.
Collapse
Affiliation(s)
- Yang Liu
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Xue Xia
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
- Macquarie Medical School, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, New South Wales, 2109, Australia
| | - Meng Zheng
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Bingyang Shi
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
- Macquarie Medical School, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, New South Wales, 2109, Australia
| |
Collapse
|
29
|
Shaw DK, Saraswathy VM, McAdow AR, Zhou L, Park D, Mote R, Johnson AN, Mokalled MH. Elevated phagocytic capacity directs innate spinal cord repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598515. [PMID: 38915507 PMCID: PMC11195157 DOI: 10.1101/2024.06.11.598515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Immune cells elicit a continuum of transcriptional and functional states after spinal cord injury (SCI). In mammals, inefficient debris clearance and chronic inflammation impede recovery and overshadow pro-regenerative immune functions. We found that, unlike mammals, zebrafish SCI elicits transient immune activation and efficient debris clearance, without causing chronic inflammation. Single-cell transcriptomics and inducible genetic ablation showed zebrafish macrophages are highly phagocytic and required for regeneration. Cross-species comparisons between zebrafish and mammalian macrophages identified transcription and immune response regulator ( tcim ) as a macrophage-enriched zebrafish gene. Genetic deletion of zebrafish tcim impairs phagocytosis and regeneration, causes aberrant and chronic immune activation, and can be rescued by transplanting wild-type immune precursors into tcim mutants. Conversely, genetic expression of human TCIM accelerates debris clearance and regeneration by reprogramming myeloid precursors into activated phagocytes. This study establishes a central requirement for elevated phagocytic capacity to achieve innate spinal cord repair.
Collapse
|
30
|
Brennan FH, Swarts EA, Kigerl KA, Mifflin KA, Guan Z, Noble BT, Wang Y, Witcher KG, Godbout JP, Popovich PG. Microglia promote maladaptive plasticity in autonomic circuitry after spinal cord injury in mice. Sci Transl Med 2024; 16:eadi3259. [PMID: 38865485 DOI: 10.1126/scitranslmed.adi3259] [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: 04/18/2023] [Accepted: 05/16/2024] [Indexed: 06/14/2024]
Abstract
Robust structural remodeling and synaptic plasticity occurs within spinal autonomic circuitry after severe high-level spinal cord injury (SCI). As a result, normally innocuous visceral or somatic stimuli elicit uncontrolled activation of spinal sympathetic reflexes that contribute to systemic disease and organ-specific pathology. How hyperexcitable sympathetic circuitry forms is unknown, but local cues from neighboring glia likely help mold these maladaptive neuronal networks. Here, we used a mouse model of SCI to show that microglia surrounded active glutamatergic interneurons and subsequently coordinated multi-segmental excitatory synaptogenesis and expansion of sympathetic networks that control immune, neuroendocrine, and cardiovascular functions. Depleting microglia during critical periods of circuit remodeling after SCI prevented maladaptive synaptic and structural plasticity in autonomic networks, decreased the frequency and severity of autonomic dysreflexia, and prevented SCI-induced immunosuppression. Forced turnover of microglia in microglia-depleted mice restored structural and functional indices of pathological dysautonomia, providing further evidence that microglia are key effectors of autonomic plasticity. Additional data show that microglia-dependent autonomic plasticity required expression of triggering receptor expressed on myeloid cells 2 (Trem2) and α2δ-1-dependent synaptogenesis. These data suggest that microglia are primary effectors of autonomic neuroplasticity and dysautonomia after SCI in mice. Manipulating microglia may be a strategy to limit autonomic complications after SCI or other forms of neurologic disease.
Collapse
Affiliation(s)
- Faith H Brennan
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Belford Center for Spinal Cord Injury, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Department of Biomedical and Molecular Sciences and Center for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Emily A Swarts
- Department of Biomedical and Molecular Sciences and Center for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Kristina A Kigerl
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Belford Center for Spinal Cord Injury, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Katherine A Mifflin
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Belford Center for Spinal Cord Injury, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Zhen Guan
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Belford Center for Spinal Cord Injury, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Benjamin T Noble
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Belford Center for Spinal Cord Injury, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Yan Wang
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Belford Center for Spinal Cord Injury, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Kristina G Witcher
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Jonathan P Godbout
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Phillip G Popovich
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Belford Center for Spinal Cord Injury, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
31
|
Li H, Watkins LR, Wang X. Microglia in neuroimmunopharmacology and drug addiction. Mol Psychiatry 2024; 29:1912-1924. [PMID: 38302560 DOI: 10.1038/s41380-024-02443-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 02/03/2024]
Abstract
Drug addiction is a chronic and debilitating disease that is considered a global health problem. Various cell types in the brain are involved in the progression of drug addiction. Recently, the xenobiotic hypothesis has been proposed, which frames substances of abuse as exogenous molecules that are responded to by the immune system as foreign "invaders", thus triggering protective inflammatory responses. An emerging body of literature reveals that microglia, the primary resident immune cells in the brain, play an important role in the progression of addiction. Repeated cycles of drug administration cause a progressive, persistent induction of neuroinflammation by releasing microglial proinflammatory cytokines and their metabolic products. This contributes to drug addiction via modulation of neuronal function. In this review, we focus on the role of microglia in the etiology of drug addiction. Then, we discuss the dynamic states of microglia and the correlative and causal evidence linking microglia to drug addiction. Finally, possible mechanisms of how microglia sense drug-related stimuli and modulate the addiction state and how microglia-targeted anti-inflammation therapies affect addiction are reviewed. Understanding the role of microglia in drug addiction may help develop new treatment strategies to fight this devastating societal challenge.
Collapse
Affiliation(s)
- Hongyuan Li
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Linda R Watkins
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China.
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China.
- Beijing National Laboratory for Molecular Sciences, Beijing, 100190, China.
| |
Collapse
|
32
|
Rutherford HA, Candeias D, Duncan CJA, Renshaw SA, Hamilton N. Macrophage transplantation rescues RNASET2-deficient leukodystrophy by replacing deficient microglia in a zebrafish model. Proc Natl Acad Sci U S A 2024; 121:e2321496121. [PMID: 38753517 PMCID: PMC11126979 DOI: 10.1073/pnas.2321496121] [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: 12/20/2023] [Accepted: 04/05/2024] [Indexed: 05/18/2024] Open
Abstract
RNASET2-deficient leukodystrophy is a rare infantile white matter disorder mimicking a viral infection and resulting in severe psychomotor impairments. Despite its severity, there is little understanding of cellular mechanisms of pathogenesis and no treatments. Recent research using the rnaset2 mutant zebrafish model has suggested that microglia may be the drivers of the neuropathology, due to their failure to digest apoptotic debris during neurodevelopment. Therefore, we developed a strategy for microglial replacement through transplantation of adult whole kidney marrow-derived macrophages into embryonic hosts. Using live imaging, we revealed that transplant-derived macrophages can engraft within host brains and express microglia-specific markers, suggesting the adoption of a microglial phenotype. Tissue-clearing strategies revealed the persistence of transplanted cells in host brains beyond embryonic stages. We demonstrated that transplanted cells clear apoptotic cells within the brain, as well as rescue overactivation of the antiviral response otherwise seen in mutant larvae. RNA sequencing at the point of peak transplant-derived cell engraftment confirms that transplantation can reduce the brain-wide immune response and particularly, the antiviral response, in rnaset2-deficient brains. Crucially, this reduction in neuroinflammation resulted in behavioral rescue-restoring rnaset2 mutant motor activity to wild-type (WT) levels in embryonic and juvenile stages. Together, these findings demonstrate the role of microglia as the cellular drivers of neuropathology in rnaset2 mutants and that macrophage transplantation is a viable strategy for microglial replacement in the zebrafish. Therefore, microglia-targeted interventions may have therapeutic benefits in RNASET2-deficient leukodystrophy.
Collapse
Affiliation(s)
- Holly A. Rutherford
- Department of Infection and Immunity, School of Medicine and Population Health, University of Sheffield, SheffieldS10 2RX, United Kingdom
- Bateson Centre, University of Sheffield, SheffieldS10 2TN, United Kingdom
| | - Diogo Candeias
- Department of Biology, University of York, YorkYO10 5DD, United Kingdom
- York Biomedical research Institute, University of York, YorkYO10 5DD, United Kingdom
| | - Christopher J. A. Duncan
- Immunology and Inflammation Theme, Translational and Clinical Research Institute, Newcastle University, NewcastleNE2 4HH, United Kingdom
- Department of Infection and Tropical Medicine, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals National Health Services Foundation Trust, NewcastleNE2 4HH, United Kingdom
| | - Stephen A. Renshaw
- Department of Infection and Immunity, School of Medicine and Population Health, University of Sheffield, SheffieldS10 2RX, United Kingdom
- Bateson Centre, University of Sheffield, SheffieldS10 2TN, United Kingdom
| | - Noémie Hamilton
- Department of Biology, University of York, YorkYO10 5DD, United Kingdom
- York Biomedical research Institute, University of York, YorkYO10 5DD, United Kingdom
| |
Collapse
|
33
|
Cao Z, Kong F, Ding J, Chen C, He F, Deng W. Promoting Alzheimer's disease research and therapy with stem cell technology. Stem Cell Res Ther 2024; 15:136. [PMID: 38715083 PMCID: PMC11077895 DOI: 10.1186/s13287-024-03737-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 04/17/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a prevalent form of dementia leading to memory loss, reduced cognitive and linguistic abilities, and decreased self-care. Current AD treatments aim to relieve symptoms and slow disease progression, but a cure is elusive due to limited understanding of the underlying disease mechanisms. MAIN CONTENT Stem cell technology has the potential to revolutionize AD research. With the ability to self-renew and differentiate into various cell types, stem cells are valuable tools for disease modeling, drug screening, and cell therapy. Recent advances have broadened our understanding beyond the deposition of amyloidβ (Aβ) or tau proteins in AD to encompass risk genes, immune system disorders, and neuron-glia mis-communication, relying heavily on stem cell-derived disease models. These stem cell-based models (e.g., organoids and microfluidic chips) simulate in vivo pathological processes with extraordinary spatial and temporal resolution. Stem cell technologies have the potential to alleviate AD pathology through various pathways, including immunomodulation, replacement of damaged neurons, and neurotrophic support. In recent years, transplantation of glial cells like oligodendrocytes and the infusion of exosomes have become hot research topics. CONCLUSION Although stem cell-based models and therapies for AD face several challenges, such as extended culture time and low differentiation efficiency, they still show considerable potential for AD treatment and are likely to become preferred tools for AD research.
Collapse
Affiliation(s)
- Zimeng Cao
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Fanshu Kong
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Jiaqi Ding
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Chunxia Chen
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China.
| | - Fumei He
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China.
- School of Pharmaceutical Sciences, Dali University, Dali, 671000, China.
| | - Wenbin Deng
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|
34
|
Mader MMD, Napole A, Wu D, Atkins M, Scavetti A, Shibuya Y, Foltz A, Hahn O, Yoo Y, Danziger R, Tan C, Wyss-Coray T, Steinman L, Wernig M. Myeloid cell replacement is neuroprotective in chronic experimental autoimmune encephalomyelitis. Nat Neurosci 2024; 27:901-912. [PMID: 38514857 DOI: 10.1038/s41593-024-01609-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/22/2024] [Indexed: 03/23/2024]
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by demyelination of the central nervous system (CNS). Autologous hematopoietic cell transplantation (HCT) shows promising benefits for relapsing-remitting MS in open-label clinical studies, but the cellular mechanisms underlying its therapeutic effects remain unclear. Using single-nucleus RNA sequencing, we identify a reactive myeloid cell state in chronic experimental autoimmune encephalitis (EAE) associated with neuroprotection and immune suppression. HCT in EAE mice results in an increase of the neuroprotective myeloid state, improvement of neurological deficits, reduced number of demyelinated lesions, decreased number of effector T cells and amelioration of reactive astrogliosis. Enhancing myeloid cell incorporation after a modified HCT further improved these neuroprotective effects. These data suggest that myeloid cell manipulation or replacement may be an effective therapeutic strategy for chronic inflammatory conditions of the CNS.
Collapse
Affiliation(s)
- Marius Marc-Daniel Mader
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Alan Napole
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Danwei Wu
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Neurology and Neurosciences, Division of Neuroimmunology and Multiple Sclerosis Center, Stanford University of Medicine, Stanford, CA, USA
| | - Micaiah Atkins
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexa Scavetti
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yohei Shibuya
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Aulden Foltz
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Oliver Hahn
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Yongjin Yoo
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ron Danziger
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Christina Tan
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Lawrence Steinman
- Department of Neurology and Neurosciences, Division of Neuroimmunology and Multiple Sclerosis Center, Stanford University of Medicine, Stanford, CA, USA
| | - Marius Wernig
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
35
|
Wang L, Zheng J, Zhao S, Wan Y, Wang M, Bosco DB, Kuan CY, Richardson JR, Wu LJ. CCR2 + monocytes replenish border-associated macrophages in the diseased mouse brain. Cell Rep 2024; 43:114120. [PMID: 38625796 PMCID: PMC11105166 DOI: 10.1016/j.celrep.2024.114120] [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: 08/05/2023] [Revised: 02/06/2024] [Accepted: 03/30/2024] [Indexed: 04/18/2024] Open
Abstract
Border-associated macrophages (BAMs) are tissue-resident macrophages that reside at the border of the central nervous system (CNS). Since BAMs originate from yolk sac progenitors that do not persist after birth, the means by which this population of cells is maintained is not well understood. Using two-photon microscopy and multiple lineage-tracing strategies, we determine that CCR2+ monocytes are significant contributors to BAM populations following disruptions of CNS homeostasis in adult mice. After BAM depletion, while the residual BAMs possess partial self-repopulation capability, the CCR2+ monocytes are a critical source of the repopulated BAMs. In addition, we demonstrate the existence of CCR2+ monocyte-derived long-lived BAMs in a brain compression model and in a sepsis model after the initial disruption of homeostasis. Our study reveals that the short-lived CCR2+ monocytes transform into long-lived BAM-like cells at the CNS border and subsequently contribute to BAM populations.
Collapse
Affiliation(s)
- Lingxiao Wang
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Shunyi Zhao
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Yushan Wan
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Meijie Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Dale B Bosco
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Chia-Yi Kuan
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Jason R Richardson
- Department of Environmental Health Science, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Center for Neuroimmunology and Glial Biology, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
36
|
Gao S, Wang Y, Li X, Liang Y, Jin Z, Yang B, Yuan TF, Tian H, Peng B, Rao Y. Dynamics of N6-methyladenosine modification during Alzheimer's disease development. Heliyon 2024; 10:e26911. [PMID: 38496847 PMCID: PMC10944207 DOI: 10.1016/j.heliyon.2024.e26911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/19/2024] Open
Abstract
N6-methyladenosine (m6A) modification is a common RNA modification in the central nervous system and has been linked to various neurological disorders, including Alzheimer's disease (AD). However, the dynamic of mRNA m6A modification and m6A enzymes during the development of AD are not well understood. Therefore, this study examined the expression profiles of m6A and its enzymes in the development of AD. The results showed that changes in the expression levels of m6A regulatory factors occur in the early stages of AD, indicating a potential role for m6A modification in the onset of the disease. Additionally, the analysis of mRNA m6A expression profiles using m6A-seq revealed significant differences in m6A modification between AD and control brains. The genes with differential methylation were found to be enriched in GO and KEGG terms related to processes such as inflammation response, immune system processes. And the differently expressed genes (DEGs) are negatively lryassociated with genes involved in microglia hemostasis, but positively associated with genes related to "disease-associated microglia" (DAM) associated genes. These findings suggest that dysregulation of mRNA m6A modification may contribute to the development of AD by affecting the function and gene expression of microglia.
Collapse
Affiliation(s)
- Shuai Gao
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, 200040, China
| | - Yuqing Wang
- Department of Neurology, Zhongshan Hospital, Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
- Department of Medical Science, Medical College of Jinzhou Medical University, Jinzhou, Liaoning, 121010, China
| | - Xiaoyu Li
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, 200040, China
| | - Yuqing Liang
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zhihao Jin
- Department of Neurology, Zhongshan Hospital, Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Baozhi Yang
- Department of Neurology, Zhongshan Hospital, Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 201108, China
| | - Hengli Tian
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Bo Peng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, 200040, China
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Yanxia Rao
- Department of Neurology, Zhongshan Hospital, Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| |
Collapse
|
37
|
Jin M, Ma Z, Dang R, Zhang H, Kim R, Xue H, Pascual J, Finkbeiner S, Head E, Liu Y, Jiang P. A Trisomy 21-linked Hematopoietic Gene Variant in Microglia Confers Resilience in Human iPSC Models of Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584646. [PMID: 38559257 PMCID: PMC10979994 DOI: 10.1101/2024.03.12.584646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
While challenging, identifying individuals displaying resilience to Alzheimer's disease (AD) and understanding the underlying mechanism holds great promise for the development of new therapeutic interventions to effectively treat AD. Down syndrome (DS), or trisomy 21, is the most common genetic cause of AD. Interestingly, some people with DS, despite developing AD neuropathology, show resilience to cognitive decline. Furthermore, DS individuals are at an increased risk of myeloid leukemia due to somatic mutations in hematopoietic cells. Recent studies indicate that somatic mutations in hematopoietic cells may lead to resilience to neurodegeneration. Microglia, derived from hematopoietic lineages, play a central role in AD etiology. We therefore hypothesize that microglia carrying the somatic mutations associated with DS myeloid leukemia may impart resilience to AD. Using CRISPR-Cas9 gene editing, we introduce a trisomy 21-linked hotspot CSF2RB A455D mutation into human pluripotent stem cell (hPSC) lines derived from both DS and healthy individuals. Employing hPSC-based in vitro microglia culture and in vivo human microglia chimeric mouse brain models, we show that in response to pathological tau, the CSF2RB A455D mutation suppresses microglial type-1 interferon signaling, independent of trisomy 21 genetic background. This mutation reduces neuroinflammation and enhances phagocytic and autophagic functions, thereby ameliorating senescent and dystrophic phenotypes in human microglia. Moreover, the CSF2RB A455D mutation promotes the development of a unique microglia subcluster with tissue repair properties. Importantly, human microglia carrying CSF2RB A455D provide protection to neuronal function, such as neurogenesis and synaptic plasticity in chimeric mouse brains where human microglia largely repopulate the hippocampus. When co-transplanted into the same mouse brains, human microglia with CSF2RB A455D mutation phagocytize and replace human microglia carrying the wildtype CSF2RB gene following pathological tau treatment. Our findings suggest that hPSC-derived CSF2RB A455D microglia could be employed to develop effective microglial replacement therapy for AD and other age-related neurodegenerative diseases, even without the need to deplete endogenous diseased microglia prior to cell transplantation.
Collapse
Affiliation(s)
- Mengmeng Jin
- Department of Cell Biology and Neuroscience, Rutgers University New Brunswick, Piscataway, NJ 08854, USA
| | - Ziyuan Ma
- Department of Cell Biology and Neuroscience, Rutgers University New Brunswick, Piscataway, NJ 08854, USA
| | - Rui Dang
- Department of Cell Biology and Neuroscience, Rutgers University New Brunswick, Piscataway, NJ 08854, USA
| | - Haiwei Zhang
- Department of Cell Biology and Neuroscience, Rutgers University New Brunswick, Piscataway, NJ 08854, USA
| | - Rachael Kim
- Department of Cell Biology and Neuroscience, Rutgers University New Brunswick, Piscataway, NJ 08854, USA
| | - Haipeng Xue
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA
| | - Jesse Pascual
- Department of Pathology and Laboratory Medicine, Department of Neurology, University of California, Irvine, CA 92697, USA
| | - Steven Finkbeiner
- Ceter for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes; University of California, San Francisco, CA 94158, USA
- Departments of Neurology and Physiology, University of California, San Francisco, CA 94158, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, Department of Neurology, University of California, Irvine, CA 92697, USA
| | - Ying Liu
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA
| | - Peng Jiang
- Department of Cell Biology and Neuroscience, Rutgers University New Brunswick, Piscataway, NJ 08854, USA
| |
Collapse
|
38
|
Li Y, Li P, Tao Q, Abuqeis IJA, Xiyang Y. Role and limitation of cell therapy in treating neurological diseases. IBRAIN 2024; 10:93-105. [PMID: 38682022 PMCID: PMC11045202 DOI: 10.1002/ibra.12152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/01/2024] [Accepted: 03/01/2024] [Indexed: 05/01/2024]
Abstract
The central role of the brain in governing systemic functions within human physiology underscores its paramount significance as the focal point of physiological regulation. The brain, a highly sophisticated organ, orchestrates a diverse array of physiological processes encompassing motor control, sensory perception, cognition, emotion, and the regulation of vital functions, such as heartbeat, respiration, and hormonal equilibrium. A notable attribute of neurological diseases manifests as the depletion of neurons and the occurrence of tissue necrosis subsequent to injury. The transplantation of neural stem cells (NSCs) into the brain exhibits the potential for the replacement of lost neurons and the reconstruction of neural circuits. Furthermore, the transplantation of other types of cells in alternative locations can secrete nutritional factors that indirectly contribute to the restoration of nervous system equilibrium and the mitigation of neural inflammation. This review summarized a comprehensive investigation into the role of NSCs, hematopoietic stem cells, mesenchymal stem cells, and support cells like astrocytes and microglia in alleviating neurological deficits after cell infusion. Moreover, a thorough assessment was undertaken to discuss extant constraints in cellular transplantation therapies, concurrently delineating indispensable model-based methodologies, specifically on organoids, which were essential for guiding prospective research initiatives in this specialized field.
Collapse
Affiliation(s)
- Yu‐Qi Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
| | - Peng‐Fei Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
| | - Qian Tao
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
| | | | - Yan‐Bin Xiyang
- School of Basic MedicineKunming Medical UniversityKunmingChina
- Department of Pharmacology and Toxicology, College of PharmacologyUniversity of ArizonaTucsonArizonaUSA
| |
Collapse
|
39
|
Rao Y, Peng B. Allogenic microglia replacement: A novel therapeutic strategy for neurological disorders. FUNDAMENTAL RESEARCH 2024; 4:237-245. [PMID: 38933508 PMCID: PMC11197774 DOI: 10.1016/j.fmre.2023.02.025] [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: 09/17/2022] [Revised: 11/17/2022] [Accepted: 02/19/2023] [Indexed: 03/29/2023] Open
Abstract
Microglia are resident immune cells in the central nervous system (CNS) that play vital roles in CNS development, homeostasis and disease pathogenesis. Genetic defects in microglia lead to microglial dysfunction, which in turn leads to neurological disorders. The correction of the specific genetic defects in microglia in these disorders can lead to therapeutic effects. Traditional genetic defect correction approaches are dependent on viral vector-based genetic defect corrections. However, the viruses used in these approaches, including adeno-associated viruses, lentiviruses and retroviruses, do not primarily target microglia; therefore, viral vector-based genetic defect corrections are ineffective in microglia. Microglia replacement is a novel approach to correct microglial genetic defects via replacing microglia of genetic defects with allogenic healthy microglia. In this paper, we systematically review the history, rationale and therapeutic perspectives of microglia replacement, which would be a novel strategy for treating CNS disorders.
Collapse
Affiliation(s)
- Yanxia Rao
- Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Bo Peng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200000, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| |
Collapse
|
40
|
Qi H, Duan S, Xu Y, Zhang H. Frontiers and future perspectives of neuroimmunology. FUNDAMENTAL RESEARCH 2024; 4:206-217. [PMID: 38933499 PMCID: PMC11197808 DOI: 10.1016/j.fmre.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 08/16/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
Neuroimmunology is an interdisciplinary branch of biomedical science that emerges from the intersection of studies on the nervous system and the immune system. The complex interplay between the two systems has long been recognized. Research efforts directed at the underlying functional interface and associated pathophysiology, however, have garnered attention only in recent decades. In this narrative review, we highlight significant advances in research on neuroimmune interplay and modulation. A particular focus is on early- and middle-career neuroimmunologists in China and their achievements in frontier areas of "neuroimmune interface", "neuro-endocrine-immune network and modulation", "neuroimmune interactions in diseases", "meningeal lymphatic and glymphatic systems in health and disease", and "tools and methodologies in neuroimmunology research". Key scientific questions and future directions for potential breakthroughs in neuroimmunology research are proposed.
Collapse
Affiliation(s)
- Hai Qi
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Shumin Duan
- Faculty of Medicine and Pharmaceutical Sciences, Zhejiang University, Hangzhou 310014, China
| | - Yanying Xu
- Department of Life Sciences, National Natural Science Foundation of China, Beijing 100085, China
| | - Hongliang Zhang
- Department of Life Sciences, National Natural Science Foundation of China, Beijing 100085, China
| |
Collapse
|
41
|
Dalmau Gasull A, Glavan M, Samawar SKR, Kapupara K, Kelk J, Rubio M, Fumagalli S, Sorokin L, Vivien D, Prinz M. The niche matters: origin, function and fate of CNS-associated macrophages during health and disease. Acta Neuropathol 2024; 147:37. [PMID: 38347231 PMCID: PMC10861620 DOI: 10.1007/s00401-023-02676-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/13/2023] [Accepted: 12/22/2023] [Indexed: 02/15/2024]
Abstract
There are several cellular and acellular structural barriers associated with the brain interfaces, which include the dura, the leptomeninges, the perivascular space and the choroid plexus epithelium. Each structure is enriched by distinct myeloid populations, which mainly originate from erythromyeloid precursors (EMP) in the embryonic yolk sac and seed the CNS during embryogenesis. However, depending on the precise microanatomical environment, resident myeloid cells differ in their marker profile, turnover and the extent to which they can be replenished by blood-derived cells. While some EMP-derived cells seed the parenchyma to become microglia, others engraft the meninges and become CNS-associated macrophages (CAMs), also referred to as border-associated macrophages (BAMs), e.g., leptomeningeal macrophages (MnMΦ). Recent data revealed that MnMΦ migrate into perivascular spaces postnatally where they differentiate into perivascular macrophages (PvMΦ). Under homeostatic conditions in pathogen-free mice, there is virtually no contribution of bone marrow-derived cells to MnMΦ and PvMΦ, but rather to macrophages of the choroid plexus and dura. In neuropathological conditions in which the blood-brain barrier is compromised, however, an influx of bone marrow-derived cells into the CNS can occur, potentially contributing to the pool of CNS myeloid cells. Simultaneously, resident CAMs may also proliferate and undergo transcriptional and proteomic changes, thereby, contributing to the disease outcome. Thus, both resident and infiltrating myeloid cells together act within their microenvironmental niche, but both populations play crucial roles in the overall disease course. Here, we summarize the current understanding of the sources and fates of resident CAMs in health and disease, and the role of the microenvironment in influencing their maintenance and function.
Collapse
Affiliation(s)
- Adrià Dalmau Gasull
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Martina Glavan
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), 14000, Caen, France
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, USA
| | - Sai K Reddy Samawar
- Institute of Physiological Chemistry and Pathobiochemistry and Cells in Motion Interfaculty Centre (CIMIC), University of Münster, Münster, Germany
| | - Kishan Kapupara
- Institute of Physiological Chemistry and Pathobiochemistry and Cells in Motion Interfaculty Centre (CIMIC), University of Münster, Münster, Germany
| | - Joe Kelk
- Laboratory of Stroke and Vascular Dysfunctions, Department of Acute Brain and Cardiovascular Injury, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, 20156, Milan, Italy
| | - Marina Rubio
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), 14000, Caen, France
| | - Stefano Fumagalli
- Laboratory of Stroke and Vascular Dysfunctions, Department of Acute Brain and Cardiovascular Injury, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, 20156, Milan, Italy
| | - Lydia Sorokin
- Institute of Physiological Chemistry and Pathobiochemistry and Cells in Motion Interfaculty Centre (CIMIC), University of Münster, Münster, Germany
| | - Denis Vivien
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), 14000, Caen, France
- Department of Clinical Research, Caen-Normandie University Hospital, CHU, Avenue de La Côte de Nacre, Caen, France
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Signalling Research Centres BIOSS and CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
42
|
Kim AB, Xiao Q, Yan P, Pan Q, Pandey G, Grathwohl S, Gonzales E, Xu I, Cho Y, Haecker H, Epelman S, Diwan A, Lee JM, DeSelm CJ. Chimeric antigen receptor macrophages target and resorb amyloid plaques. JCI Insight 2024; 9:e175015. [PMID: 38516884 PMCID: PMC11063938 DOI: 10.1172/jci.insight.175015] [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: 08/21/2023] [Accepted: 01/31/2024] [Indexed: 03/23/2024] Open
Abstract
Substantial evidence suggests a role for immunotherapy in treating Alzheimer's disease (AD). While the precise pathophysiology of AD is incompletely understood, clinical trials of antibodies targeting aggregated forms of β amyloid (Aβ) have shown that reducing amyloid plaques can mitigate cognitive decline in patients with early-stage AD. Here, we describe what we believe to be a novel approach to target and degrade amyloid plaques by genetically engineering macrophages to express an Aβ-targeting chimeric antigen receptor (CAR-Ms). When injected intrahippocampally, first-generation CAR-Ms have limited persistence and fail to significantly reduce plaque load, which led us to engineer next-generation CAR-Ms that secrete M-CSF and self-maintain without exogenous cytokines. Cytokine secreting "reinforced CAR-Ms" have greater survival in the brain niche and significantly reduce plaque load locally in vivo. These findings support CAR-Ms as a platform to rationally target, resorb, and degrade pathogenic material that accumulates with age, as exemplified by targeting Aβ in AD.
Collapse
Affiliation(s)
- Alexander B. Kim
- Department of Radiation Oncology
- Bursky Center for Human Immunology and Immunotherapy
| | - Qingli Xiao
- Department of Neurology, and
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ping Yan
- Department of Neurology, and
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Qiuyun Pan
- Department of Radiation Oncology
- Bursky Center for Human Immunology and Immunotherapy
| | - Gaurav Pandey
- Department of Radiation Oncology
- Bursky Center for Human Immunology and Immunotherapy
| | - Susie Grathwohl
- Department of Neurology, and
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ernesto Gonzales
- Department of Neurology, and
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Isabella Xu
- Department of Neurology, and
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yoonho Cho
- Department of Neurology, and
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Hans Haecker
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Slava Epelman
- Department of Medicine, Division of Cardiology, Peter Munk Cardiac Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Abhinav Diwan
- Department of Neurology, and
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
- Departments of Medicine, Cell Biology and Physiology, Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri, USA
- Medicine Service, St. Louis VA Medical Center, St. Louis, Missouri, USA
| | - Jin-Moo Lee
- Department of Neurology, and
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Carl J. DeSelm
- Department of Radiation Oncology
- Bursky Center for Human Immunology and Immunotherapy
| |
Collapse
|
43
|
Zhang L, Li G, Li Y. TRIM59 suppresses the brain ischaemia/reperfusion injury and pyroptosis of microglial through mediating the ubiquitination of NLRP3. Sci Rep 2024; 14:2511. [PMID: 38291200 PMCID: PMC10828378 DOI: 10.1038/s41598-024-52914-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/25/2024] [Indexed: 02/01/2024] Open
Abstract
Cerebral ischaemia/reperfusion (I/R) injury induces irreversible brain injury and causes functional impairment. Ubiquitination plays a crucial role in protein degradation, but its role in cerebral I/R injury remains unclear. Differentially expressed genes in stroke were identified by analysing the microarray dataset GSE119121. Cerebral I/R was simulated in vitro by treating human microglial HMC3 cells with oxygen-glucose deprivation/reperfusion (OGD/R). Cell viability was tested by Cell Counting Kit 8 (CCK-8) assays, and pyroptosis was examined by flow cytometry. Lactate dehydrogenase (LDH) and inflammatory cytokine secretion were measured by LDH cytotoxicity assays and enzyme-linked immunosorbent assay (ELISA), respectively. The cerebral I/R animal model was established by middle cerebral artery occlusion (MCAO) surgery in rats. Bioinformatic analysis indicated that tripartite motif-containing protein 59 (TRIM59) is downregulated in stroke, which was verified in cerebral I/R models. The upregulation of TRIM59 promoted viability and inhibited pyroptosis in OGD/R-treated microglia and alleviated cerebral I/R injury in vivo. TRIM59 attenuated NOD-like receptor family pyrin domain containing 3 (NLRP3) protein expression through ubiquitination, thus degrading NLRP3 and alleviating OGD/R-induced injury. TRIM59 relieves cerebral I/R injury in vivo and in vivo. Mechanistically, TRIM59 directly interacts with NLRP3 and inhibits NLRP3 through ubiquitination. Targeting the TRIM59/NLRP3 signalling axis may be an effective therapeutic strategy for cerebral I/R.
Collapse
Affiliation(s)
- Liangtian Zhang
- Department of Emergency Medicine, Chun'an First People's Hospital, Hangzhou City, Zhejiang Province, China
| | - Gang Li
- Emergency and Critical Care Center, Department of Emergency Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Ying Li
- Department of Special Inspection, Hangzhou TCM Hospital, Affiliated to Zhejiang Chinese Medical University, No. 453, Tiyuchang Road, Hangzhou City, Zhejiang Province, China.
| |
Collapse
|
44
|
Khan R, Ritzel RM. Maestros of anesthesia. eLife 2024; 13:e95064. [PMID: 38236208 PMCID: PMC10796144 DOI: 10.7554/elife.95064] [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: 01/19/2024] Open
Abstract
Microglia regulate anesthesia by altering the activity of neurons in specific regions of the brain via a purinergic receptor.
Collapse
Affiliation(s)
- Romeesa Khan
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical SciencesHoustonUnited States
- Department of Neurology, McGovern Medical School, University of TexasHoustonUnited States
| | - Rodney M Ritzel
- Department of Neurology, McGovern Medical School, University of TexasHoustonUnited States
| |
Collapse
|
45
|
Weyer MP, Strehle J, Schäfer MKE, Tegeder I. Repurposing of pexidartinib for microglia depletion and renewal. Pharmacol Ther 2024; 253:108565. [PMID: 38052308 DOI: 10.1016/j.pharmthera.2023.108565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/07/2023]
Abstract
Pexidartinib (PLX3397) is a small molecule receptor tyrosine kinase inhibitor of colony stimulating factor 1 receptor (CSF1R) with moderate selectivity over other members of the platelet derived growth factor receptor family. It is approved for treatment of tenosynovial giant cell tumors (TGCT). CSF1R is highly expressed by microglia, which are macrophages of the central nervous system (CNS) that defend the CNS against injury and pathogens and contribute to synapse development and plasticity. Challenged by pathogens, apoptotic cells, debris, or inflammatory molecules they adopt a responsive state to propagate the inflammation and eventually return to a homeostatic state. The phenotypic switch may fail, and disease-associated microglia contribute to the pathophysiology in neurodegenerative or neuropsychiatric diseases or long-lasting detrimental brain inflammation after brain, spinal cord or nerve injury or ischemia/hemorrhage. Microglia also contribute to the growth permissive tumor microenvironment of glioblastoma (GBM). In rodents, continuous treatment for 1-2 weeks via pexidartinib food pellets leads to a depletion of microglia and subsequent repopulation from the remaining fraction, which is aided by peripheral monocytes that search empty niches for engraftment. The putative therapeutic benefit of such microglia depletion or forced renewal has been assessed in almost any rodent model of CNS disease or injury or GBM with heterogeneous outcomes, but a tendency of partial beneficial effects. So far, microglia monitoring e.g. via positron emission imaging is not standard of care for patients receiving Pexidartinib (e.g. for TGCT), so that the depletion and repopulation efficiency in humans is still largely unknown. Considering the virtuous functions of microglia, continuous depletion is likely no therapeutic option but short-lasting transient partial depletion to stimulate microglia renewal or replace microglia in genetic disease in combination with e.g. stem cell transplantation or as part of a multimodal concept in treatment of glioblastoma appears feasible. The present review provides an overview of the preclinical evidence pro and contra microglia depletion as a therapeutic approach.
Collapse
Affiliation(s)
- Marc-Philipp Weyer
- Institute of Clinical Pharmacology, Goethe-University Frankfurt, Faculty of Medicine, Frankfurt, Germany
| | - Jenny Strehle
- Department of Anesthesiology, University Medical Center Johannes Gutenberg-University Mainz, Germany
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center Johannes Gutenberg-University Mainz, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Goethe-University Frankfurt, Faculty of Medicine, Frankfurt, Germany.
| |
Collapse
|
46
|
Sankowski R, Süß P, Benkendorff A, Böttcher C, Fernandez-Zapata C, Chhatbar C, Cahueau J, Monaco G, Gasull AD, Khavaran A, Grauvogel J, Scheiwe C, Shah MJ, Heiland DH, Schnell O, Markfeld-Erol F, Kunze M, Zeiser R, Priller J, Prinz M. Multiomic spatial landscape of innate immune cells at human central nervous system borders. Nat Med 2024; 30:186-198. [PMID: 38123840 PMCID: PMC10803260 DOI: 10.1038/s41591-023-02673-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/30/2023] [Indexed: 12/23/2023]
Abstract
The innate immune compartment of the human central nervous system (CNS) is highly diverse and includes several immune-cell populations such as macrophages that are frequent in the brain parenchyma (microglia) and less numerous at the brain interfaces as CNS-associated macrophages (CAMs). Due to their scantiness and particular location, little is known about the presence of temporally and spatially restricted CAM subclasses during development, health and perturbation. Here we combined single-cell RNA sequencing, time-of-flight mass cytometry and single-cell spatial transcriptomics with fate mapping and advanced immunohistochemistry to comprehensively characterize the immune system at human CNS interfaces with over 356,000 analyzed transcriptomes from 102 individuals. We also provide a comprehensive analysis of resident and engrafted myeloid cells in the brains of 15 individuals with peripheral blood stem cell transplantation, revealing compartment-specific engraftment rates across different CNS interfaces. Integrated multiomic and high-resolution spatial transcriptome analysis of anatomically dissected glioblastoma samples shows regionally distinct myeloid cell-type distributions driven by hypoxia. Notably, the glioblastoma-associated hypoxia response was distinct from the physiological hypoxia response in fetal microglia and CAMs. Our results highlight myeloid diversity at the interfaces of the human CNS with the periphery and provide insights into the complexities of the human brain's immune system.
Collapse
Affiliation(s)
- Roman Sankowski
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Patrick Süß
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Molecular Neurology, Friedrich Alexander University Erlangen-Nürnberg, University Hospital Erlangen, Erlangen, Germany
| | - Alexander Benkendorff
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Chotima Böttcher
- Neuropsychiatry Unit and Laboratory of Molecular Psychiatry, Charité, Universitätsmedizin Berlin and DZNE, Berlin, Germany
| | - Camila Fernandez-Zapata
- Neuropsychiatry Unit and Laboratory of Molecular Psychiatry, Charité, Universitätsmedizin Berlin and DZNE, Berlin, Germany
| | - Chintan Chhatbar
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jonathan Cahueau
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gianni Monaco
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg, Germany
| | - Adrià Dalmau Gasull
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ashkan Khavaran
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jürgen Grauvogel
- Department of Neurosurgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Scheiwe
- Department of Neurosurgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Mukesch Johannes Shah
- Department of Neurosurgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dieter Henrik Heiland
- Department of Neurosurgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Oliver Schnell
- Department of Neurosurgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Filiz Markfeld-Erol
- Department of Gynecology, Obstetrics, and Perinatology, Faculty of Medicine, University Hospital, Freiburg, Germany
| | - Mirjam Kunze
- Department of Gynecology, Obstetrics, and Perinatology, Faculty of Medicine, University Hospital, Freiburg, Germany
| | - Robert Zeiser
- Department of Internal Medicine I, Faculty of Medicine, Medical Center-University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Josef Priller
- Neuropsychiatry Unit and Laboratory of Molecular Psychiatry, Charité, Universitätsmedizin Berlin and DZNE, Berlin, Germany
- Department of Psychiatry and Psychotherapy, School of Medicine and Health, Technical University of Munich, Munich, Germany
- University of Edinburgh and UK DRI, Edinburgh, UK
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
47
|
He Y, Liu T, He Q, Ke W, Li X, Du J, Deng S, Shu Z, Wu J, Yang B, Wang Y, Mao Y, Rao Y, Shu Y, Peng B. Microglia facilitate and stabilize the response to general anesthesia via modulating the neuronal network in a brain region-specific manner. eLife 2023; 12:RP92252. [PMID: 38131301 PMCID: PMC10746144 DOI: 10.7554/elife.92252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
General anesthesia leads to a loss of consciousness and an unrousable state in patients. Although general anesthetics are widely used in clinical practice, their underlying mechanisms remain elusive. The potential involvement of nonneuronal cells is unknown. Microglia are important immune cells in the central nervous system (CNS) that play critical roles in CNS function and dysfunction. We unintentionally observed delayed anesthesia induction and early anesthesia emergence in microglia-depleted mice. We found that microglial depletion differentially regulates neuronal activities by suppressing the neuronal network of anesthesia-activated brain regions and activating emergence-activated brain regions. Thus, microglia facilitate and stabilize the anesthesia status. This influence is not mediated by dendritic spine plasticity. Instead, it relies on the activation of microglial P2Y12 and subsequent calcium influx, which facilitates the general anesthesia response. Together, we elucidate the regulatory role of microglia in general anesthesia, extending our knowledge of how nonneuronal cells modulate neuronal activities.
Collapse
Affiliation(s)
- Yang He
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Taohui Liu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Quansheng He
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Wei Ke
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Xiaoyu Li
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Jinjin Du
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
- School of Basic Medical Sciences, Jinzhou Medical UniversityJinzhouChina
| | - Suixin Deng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Zhenfeng Shu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Jialin Wu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Baozhi Yang
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
- School of Basic Medical Sciences, Jinzhou Medical UniversityJinzhouChina
| | - Yuqing Wang
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
- School of Basic Medical Sciences, Jinzhou Medical UniversityJinzhouChina
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Yanxia Rao
- Department of Neurology, Zhongshan Hospital, Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan UniversityShanghaiChina
| | - Yousheng Shu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Bo Peng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
- Co-Innovation Center of Neurodegeneration, Nantong UniversityNantongChina
| |
Collapse
|
48
|
Zhu H, Guan A, Liu J, Peng L, Zhang Z, Wang S. Noteworthy perspectives on microglia in neuropsychiatric disorders. J Neuroinflammation 2023; 20:223. [PMID: 37794488 PMCID: PMC10548593 DOI: 10.1186/s12974-023-02901-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 09/22/2023] [Indexed: 10/06/2023] Open
Abstract
Microglia are so versatile that they not only provide immune surveillance for central nervous system, but participate in neural circuitry development, brain blood vessels formation, blood-brain barrier architecture, and intriguingly, the regulation of emotions and behaviors. Microglia have a profound impact on neuronal survival, brain wiring and synaptic plasticity. As professional phagocytic cells in the brain, they remove dead cell debris and neurotoxic agents via an elaborate mechanism. The functional profile of microglia varies considerately depending on age, gender, disease context and other internal or external environmental factors. Numerous studies have demonstrated a pivotal involvement of microglia in neuropsychiatric disorders, including negative affection, social deficit, compulsive behavior, fear memory, pain and other symptoms associated with major depression disorder, anxiety disorder, autism spectrum disorder and schizophrenia. In this review, we summarized the latest discoveries regarding microglial ontogeny, cell subtypes or state spectrum, biological functions and mechanistic underpinnings of emotional and behavioral disorders. Furthermore, we highlight the potential of microglia-targeted therapies of neuropsychiatric disorders, and propose outstanding questions to be addressed in future research of human microglia.
Collapse
Affiliation(s)
- Hongrui Zhu
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Ao Guan
- School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Jiayuan Liu
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Li Peng
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Zhi Zhang
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
- Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Sheng Wang
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| |
Collapse
|
49
|
Dissecting microglial aging and creating a model of aged microglia in a non-aged brain. NATURE AGING 2023; 3:1185-1186. [PMID: 37697167 DOI: 10.1038/s43587-023-00487-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
|
50
|
Li X, Li Y, Jin Y, Zhang Y, Wu J, Xu Z, Huang Y, Cai L, Gao S, Liu T, Zeng F, Wang Y, Wang W, Yuan TF, Tian H, Shu Y, Guo F, Lu W, Mao Y, Mei X, Rao Y, Peng B. Transcriptional and epigenetic decoding of the microglial aging process. NATURE AGING 2023; 3:1288-1311. [PMID: 37697166 PMCID: PMC10570141 DOI: 10.1038/s43587-023-00479-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 08/03/2023] [Indexed: 09/13/2023]
Abstract
As important immune cells, microglia undergo a series of alterations during aging that increase the susceptibility to brain dysfunctions. However, the longitudinal characteristics of microglia remain poorly understood. In this study, we mapped the transcriptional and epigenetic profiles of microglia from 3- to 24-month-old mice. We first discovered unexpected sex differences and identified age-dependent microglia (ADEM) genes during the aging process. We then compared the features of aging and reactivity in female microglia at single-cell resolution and epigenetic level. To dissect functions of aged microglia excluding the influence from other aged brain cells, we established an accelerated microglial turnover model without directly affecting other brain cells. By this model, we achieved aged-like microglia in non-aged brains and confirmed that aged-like microglia per se contribute to cognitive decline. Collectively, our work provides a comprehensive resource for decoding the aging process of microglia, shedding light on how microglia maintain brain functions.
Collapse
Affiliation(s)
- Xiaoyu Li
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Yuxin Li
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Yuxiao Jin
- Department of Neurology, Zhongshan Hospital, Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yuheng Zhang
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Jingchuan Wu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhen Xu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yubin Huang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Lin Cai
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuai Gao
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Taohui Liu
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Fanzhuo Zeng
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yafei Wang
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Wenxu Wang
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hengli Tian
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yousheng Shu
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Feifan Guo
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Wei Lu
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Xifan Mei
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yanxia Rao
- Department of Neurology, Zhongshan Hospital, Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| | - Bo Peng
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, China.
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.
- Co-Innovation Center of Neurodegeneration, Nantong University, Nantong, China.
| |
Collapse
|