51
|
Lloyd AF, Martinez-Muriana A, Davis E, Daniels MJD, Hou P, Mancuso R, Brenes AJ, Sinclair LV, Geric I, Snellinx A, Craessaerts K, Theys T, Fiers M, De Strooper B, Howden AJM. Deep proteomic analysis of microglia reveals fundamental biological differences between model systems. Cell Rep 2024; 43:114908. [PMID: 39460937 DOI: 10.1016/j.celrep.2024.114908] [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: 05/16/2024] [Revised: 09/06/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Using high-resolution quantitative mass spectrometry, we present comprehensive human and mouse microglia proteomic datasets consisting of over 11,000 proteins across six microglia groups. Microglia share a core protein signature of over 5,600 proteins, yet fundamental differences are observed between species and culture conditions. Mouse microglia demonstrate proteome differences in inflammation- and Alzheimer's disease-associated proteins. We identify differences in the protein content of ex vivo and in vitro cells and significant proteome differences associated with protein synthesis, metabolism, microglia marker expression, and environmental sensors. Culturing microglia induces rapidly increased growth, protein content, and inflammatory protein expression. These changes are restored by engrafting in vitro cells into the brain, with xenografted human embryonic stem cell (hESC)-derived microglia closely resembling microglia from the human brain. These data provide an important resource for the field and highlight important considerations needed when using model systems to study human physiology and pathology of microglia.
Collapse
Affiliation(s)
- Amy F Lloyd
- Cell Signaling and Immunology, University of Dundee, Dundee, UK.
| | - Anna Martinez-Muriana
- VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Emma Davis
- The Francis Crick Institute, London, UK; UK Dementia Research Institute at UCL, University College London, London, UK
| | | | - Pengfei Hou
- VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Renzo Mancuso
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; MINDlab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Alejandro J Brenes
- Cell Signaling and Immunology, University of Dundee, Dundee, UK; Centre for Gene Regulation and Expression, University of Dundee, Dundee, UK
| | | | - Ivana Geric
- VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - An Snellinx
- VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Katleen Craessaerts
- VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Tom Theys
- Department of Neurosciences, Research Group Experimental Neurosurgery and Neuroanatomy, KU Leuven, Leuven, Belgium
| | - Mark Fiers
- VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Bart De Strooper
- VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium; The Francis Crick Institute, London, UK; UK Dementia Research Institute at UCL, University College London, London, UK.
| | | |
Collapse
|
52
|
Balak CD, Schlachetzki JCM, Lana AJ, West E, Hong C, DuGal J, Zhou Y, Li B, Saisan P, Spann NJ, Sarsani V, Pasillas MP, O'Brien S, Gordts P, Stevens B, Kamme F, Glass CK. Mechanisms driving epigenetic and transcriptional responses of microglia in a neurodegenerative lysosomal storage disorder model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.12.623296. [PMID: 39605454 PMCID: PMC11601307 DOI: 10.1101/2024.11.12.623296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Lysosomal dysfunction is causally linked to neurodegeneration in many lysosomal storage disorders (LSDs) and is associated with various age-related neurodegenerative diseases 1,2 , but there is limited understanding of the mechanisms by which altered lysosomal function leads to changes in gene expression that drive pathogenic cellular phenotypes. To investigate this question, we performed systematic imaging, transcriptomic, and epigenetic studies of major brain cell types in Sgsh null (KO) mice, a preclinical mouse model for Sanfilippo syndrome (Mucopolysaccharidosis Type IIIA, MPS-IIIA) 3,4 . MPS-IIIA is a neurodegenerative LSD caused by homozygous loss-of-function (LoF) mutations in SGSH which results in severe early-onset developmental, behavioral, and neurocognitive impairment 5-15 . Electron microscopy, immunohistochemistry, and single-nucleus RNA-sequencing analysis revealed microglia as the cell type exhibiting the most dramatic phenotypic alterations in Sgsh KO mice. Further temporal analysis of microglia gene expression showed dysregulation of genes associated with lysosomal function and immune signaling pathways beginning early in the course of the disease. Sgsh deficiency similarly resulted in increases in open chromatin and histone acetylation at thousands of putative microglia-specific enhancers associated with upregulated genes but had much less impact on the epigenetic landscapes of neurons or oligodendrocytes. We provide evidence for dominant and context-dependent roles of members of the MITF/TFE family as major drivers of microglia-specific epigenetic and transcriptional changes resulting from lysosomal stress that are dependent on collaborative interactions with PU.1/ETS and C/EBP transcription factors. Lastly, we show that features of the transcriptomic and epigenetic alterations observed in murine Sgsh deficiency are also observed in microglia derived from mouse models of age-related neurodegeneration and in human Alzheimer's disease patients, revealing common and disease-specific transcriptional mechanisms associated with disease-associated microglia phenotypes.
Collapse
|
53
|
Ma W, Zhao L, Xu B, Fariss RN, Redmond TM, Zou J, Wong WT, Li W. Human-induced pluripotent stem cell-derived microglia integrate into mouse retina and recapitulate features of endogenous microglia. eLife 2024; 12:RP90695. [PMID: 39514271 PMCID: PMC11587526 DOI: 10.7554/elife.90695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Microglia exhibit both maladaptive and adaptive roles in the pathogenesis of neurodegenerative diseases and have emerged as a cellular target for central nervous system (CNS) disorders, including those affecting the retina. Replacing maladaptive microglia, such as those impacted by aging or over-activation, with exogenous microglia that can enable adaptive functions has been proposed as a potential therapeutic strategy for neurodegenerative diseases. To investigate microglia replacement as an approach for retinal diseases, we first employed a protocol to efficiently generate human-induced pluripotent stem cell (hiPSC)-derived microglia in quantities sufficient for in vivo transplantation. These cells demonstrated expression of microglia-enriched genes and showed typical microglial functions such as LPS-induced responses and phagocytosis. We then performed xenotransplantation of these hiPSC-derived microglia into the subretinal space of adult mice whose endogenous retinal microglia have been pharmacologically depleted. Long-term analysis post-transplantation demonstrated that transplanted hiPSC-derived microglia successfully integrated into the neuroretina as ramified cells, occupying positions previously filled by the endogenous microglia and expressed microglia homeostatic markers such as P2ry12 and Tmem119. Furthermore, these cells were found juxtaposed alongside residual endogenous murine microglia for up to 8 months in the retina, indicating their ability to establish a stable homeostatic state in vivo. Following retinal pigment epithelial cell injury, transplanted microglia demonstrated responses typical of endogenous microglia, including migration, proliferation, and phagocytosis. Our findings indicate the feasibility of microglial transplantation and integration in the retina and suggest that modulating microglia through replacement may be a therapeutic strategy for treating neurodegenerative retinal diseases.
Collapse
Affiliation(s)
- Wenxin Ma
- Retinal Neurophysiology Section, National Eye InstituteBethesdaUnited States
| | - Lian Zhao
- Genetic Engineering Core, National Eye InstituteBethesdaUnited States
| | - Biying Xu
- Immunoregulation Section, National Eye InstituteBethesdaUnited States
| | - Robert N Fariss
- Biological Imaging Core, National Eye InstituteBethesdaUnited States
| | - T Michael Redmond
- Molecular Mechanisms Section, National Eye InstituteBethesdaUnited States
| | - Jizhong Zou
- iPSC Core, National Heart, Lung, and Blood InstituteBethesdaUnited States
| | | | - Wei Li
- Retinal Neurophysiology Section, National Eye InstituteBethesdaUnited States
| |
Collapse
|
54
|
Li X, Liu J, Boreland AJ, Kapadia S, Zhang S, Stillitano AC, Abbo Y, Clark L, Lai D, Liu Y, Barr PB, Meyers JL, Kamarajan C, Kuang W, Agrawal A, Slesinger PA, Dick D, Salvatore J, Tischfield J, Duan J, Edenberg HJ, Kreimer A, Hart RP, Pang ZP. Polygenic risk for alcohol use disorder affects cellular responses to ethanol exposure in a human microglial cell model. SCIENCE ADVANCES 2024; 10:eado5820. [PMID: 39514655 PMCID: PMC11546823 DOI: 10.1126/sciadv.ado5820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024]
Abstract
Polygenic risk scores (PRSs) assess genetic susceptibility to alcohol use disorder (AUD), yet their molecular implications remain underexplored. Neuroimmune interactions, particularly in microglia, are recognized as notable contributors to AUD pathophysiology. We investigated the interplay between AUD PRS and ethanol in human microglia derived from iPSCs from individuals with AUD high-PRS (diagnosed with AUD) or low-PRS (unaffected). Ethanol exposure induced elevated CD68 expression and morphological changes in microglia, with differential responses between high-PRS and low-PRS microglial cells. Transcriptomic analysis revealed expression differences in MHCII complex and phagocytosis-related genes following ethanol exposure; high-PRS microglial cells displayed enhanced phagocytosis and increased CLEC7A expression, unlike low-PRS microglial cells. Synapse numbers in cocultures of induced neurons with microglia after alcohol exposure were lower in high-RPS cocultures, suggesting possible excess synapse pruning. This study provides insights into the intricate relationship between AUD PRS, ethanol, and microglial function, potentially influencing neuronal functions in developing AUD.
Collapse
Affiliation(s)
- Xindi Li
- Department of Neuroscience and Cell Biology and The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Jiayi Liu
- Department of Biochemistry and Molecular Biology, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, 679 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Andrew J. Boreland
- Department of Neuroscience and Cell Biology and The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Sneha Kapadia
- Department of Neuroscience and Cell Biology and The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Siwei Zhang
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| | - Alessandro C. Stillitano
- Department of Neuroscience and Cell Biology and The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Yara Abbo
- Department of Neuroscience and Cell Biology and The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Lorraine Clark
- Department of Neuroscience and Cell Biology and The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Dongbing Lai
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yunlong Liu
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Peter B. Barr
- Department of Psychiatry & Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Jacquelyn L. Meyers
- Department of Psychiatry & Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Chella Kamarajan
- Department of Psychiatry & Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Weipeng Kuang
- Department of Psychiatry & Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Arpana Agrawal
- Department of Psychiatry, Washinton University School of Medicine, Saint Louis, MO 63108, USA
| | - Paul A. Slesinger
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Danielle Dick
- Department of Psychiatry, Rutgers University Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Jessica Salvatore
- Department of Psychiatry, Rutgers University Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Jay Tischfield
- Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 08854, USA
- Department of Genetics, Rutgers University, Piscataway, NJ 08854, USA
| | - Jubao Duan
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| | - Howard J. Edenberg
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Anat Kreimer
- Department of Biochemistry and Molecular Biology, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, 679 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Ronald P. Hart
- Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 08854, USA
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Zhiping P. Pang
- Department of Neuroscience and Cell Biology and The Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| |
Collapse
|
55
|
Franza M, Varricchio R, Alloisio G, De Simone G, Di Bella S, Ascenzi P, di Masi A. Zebrafish ( Danio rerio) as a Model System to Investigate the Role of the Innate Immune Response in Human Infectious Diseases. Int J Mol Sci 2024; 25:12008. [PMID: 39596075 PMCID: PMC11593600 DOI: 10.3390/ijms252212008] [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: 10/07/2024] [Revised: 10/30/2024] [Accepted: 11/03/2024] [Indexed: 11/28/2024] Open
Abstract
The zebrafish (Danio rerio) has emerged as a valuable model for studying host-pathogen interactions due to its unique combination of characteristics. These include extensive sequence and functional conservation with the human genome, optical transparency in larvae that allows for high-resolution visualization of host cell-microbe interactions, a fully sequenced and annotated genome, advanced forward and reverse genetic tools, and suitability for chemical screening studies. Despite anatomical differences with humans, the zebrafish model has proven instrumental in investigating immune responses and human infectious diseases. Notably, zebrafish larvae rely exclusively on innate immune responses during the early stages of development, as the adaptive immune system becomes fully functional only after 4-6 weeks post-fertilization. This window provides a unique opportunity to isolate and examine infection and inflammation mechanisms driven by the innate immune response without the confounding effects of adaptive immunity. In this review, we highlight the strengths and limitations of using zebrafish as a powerful vertebrate model to study innate immune responses in infectious diseases. We will particularly focus on host-pathogen interactions in human infections caused by various bacteria (Clostridioides difficile, Staphylococcus aureus, and Pseudomonas aeruginosa), viruses (herpes simplex virus 1, SARS-CoV-2), and fungi (Aspergillus fumigatus and Candida albicans).
Collapse
Affiliation(s)
- Maria Franza
- Department of Sciences, Roma Tre University, 00146 Roma, Italy; (M.F.); (R.V.); (G.A.); (G.D.S.); (P.A.)
| | - Romualdo Varricchio
- Department of Sciences, Roma Tre University, 00146 Roma, Italy; (M.F.); (R.V.); (G.A.); (G.D.S.); (P.A.)
| | - Giulia Alloisio
- Department of Sciences, Roma Tre University, 00146 Roma, Italy; (M.F.); (R.V.); (G.A.); (G.D.S.); (P.A.)
| | - Giovanna De Simone
- Department of Sciences, Roma Tre University, 00146 Roma, Italy; (M.F.); (R.V.); (G.A.); (G.D.S.); (P.A.)
| | - Stefano Di Bella
- Clinical Department of Medical, Surgical and Health Sciences, Trieste University, 34127 Trieste, Italy;
| | - Paolo Ascenzi
- Department of Sciences, Roma Tre University, 00146 Roma, Italy; (M.F.); (R.V.); (G.A.); (G.D.S.); (P.A.)
- Accademia Nazionale dei Lincei, 00165 Roma, Italy
| | - Alessandra di Masi
- Department of Sciences, Roma Tre University, 00146 Roma, Italy; (M.F.); (R.V.); (G.A.); (G.D.S.); (P.A.)
- Centro Linceo Interdisciplinare “Beniamino Segre”, Accademia Nazionale dei Lincei, 00165 Roma, Italy
| |
Collapse
|
56
|
Sangineto M, Ciarnelli M, Moola A, Naik Bukke V, Cassano T, Villani R, Romano AD, Di Gioia G, Avolio C, Serviddio G. Krebs cycle derivatives, dimethyl fumarate and itaconate, control metabolic reprogramming in inflammatory human microglia cell line. Mitochondrion 2024; 79:101966. [PMID: 39276907 DOI: 10.1016/j.mito.2024.101966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 08/29/2024] [Accepted: 09/08/2024] [Indexed: 09/17/2024]
Abstract
Metabolic reprogramming drives inflammatory activity in macrophages, including microglia, with Krebs cycle (KC) intermediates playing a crucial role as signaling molecules. Here, we show that the bioenergetic profile of LPS-activated human microglialclone 3 cell line (HMC3) is characterized by high levels of glycolysis and mitochondrial (mt) respiration, and the treatment with KC derivatives, namely dimethyl-fumarate (DMF) and itaconate (ITA), almost restores normal metabolism. However, despite comparable bioenergetic and anti-inflammatory effects, the mt hyper-activity was differentially modulated by DMF and ITA. DMF normalized complex I activity, while ITA dampened both complex I and II hyper-activity counteracting oxidative stress more efficiently.
Collapse
Affiliation(s)
- Moris Sangineto
- Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.
| | - Martina Ciarnelli
- Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Archana Moola
- Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Vidyasagar Naik Bukke
- Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Tommaso Cassano
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Rosanna Villani
- Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Antonino D Romano
- Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Giuseppe Di Gioia
- Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Carlo Avolio
- Neurology Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Gaetano Serviddio
- Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
57
|
Rosete C, Ciernia AV. The Two Faces of HDAC3: Neuroinflammation in Disease and Neuroprotection in Recovery. Epigenomics 2024; 16:1373-1388. [PMID: 39513228 PMCID: PMC11728336 DOI: 10.1080/17501911.2024.2419357] [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: 08/21/2024] [Accepted: 10/17/2024] [Indexed: 11/15/2024] Open
Abstract
Histone deacetylase 3 (HDAC3) is a critical regulator of gene expression, influencing a variety of cellular processes in the central nervous system. As such, dysfunction of this enzyme may serve as a key driver in the pathophysiology of various neuropsychiatric disorders and neurodegenerative diseases. HDAC3 plays a crucial role in regulating neuroinflammation, and is now widely recognized as a major contributor to neurological conditions, as well as in promoting neuroprotective recovery following brain injury, hemorrhage and stroke. Emerging evidence suggests that pharmacological inhibition of HDAC3 can mitigate behavioral and neuroimmune deficits in various brain diseases and disorders, offering a promising therapeutic strategy. Understanding HDAC3 in the healthy brain lays the necessary foundation to define and resolve its dysfunction in a disease state. This review explores the mechanisms of HDAC3 in various cell types and its involvement in disease pathology, emphasizing the potential of HDAC3 inhibition to address neuroimmune, gene expression and behavioral deficits in a range of neurodegenerative and neuropsychiatric conditions.
Collapse
Affiliation(s)
- Cal Rosete
- Djavad Mowafaghian Centre for Brain Health, Vancouver, V6T 1Z3, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, V6T 2A1, Canada
| | - Annie Vogel Ciernia
- Djavad Mowafaghian Centre for Brain Health, Vancouver, V6T 1Z3, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, V6T 2A1, Canada
| |
Collapse
|
58
|
Podlesny-Drabiniok A, Romero-Molina C, Patel T, See WY, Liu Y, Marcora E, Goate AM. Cytokine-induced reprogramming of human macrophages toward Alzheimer's disease-relevant molecular and cellular phenotypes in vitro. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.24.619910. [PMID: 39554174 PMCID: PMC11565805 DOI: 10.1101/2024.10.24.619910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Myeloid cells including brain-resident (microglia) and peripheral macrophages play a crucial role in various pathological conditions, including neurodegenerative disorders like Alzheimer's disease (AD). They respond to disruption of tissue homeostasis associated with disease conditions by acquiring various transcriptional and functional states. Experimental investigation of these states is hampered by the lack of tools that enable accessible and robust reprogramming of human macrophages toward Alzheimer's disease-relevant molecular and cellular phenotypes in vitro. In this study, we investigated the ability of a cytokine mix, including interleukin-4 (IL4), colony stimulating factor 1 (CSF1/MCSF), interleukin 34 (IL34) and transforming growth factor beta (TGFβ), to induce reprogramming of cultured human THP-1 macrophages. Our results indicate this treatment led to significant transcriptomic changes, driving THP-1 macrophages towards a transcriptional state reminiscent of disease-associated microglia (DAM) and lipid-associated macrophages (LAM) collectively referred to as DLAM. Transcriptome profiling revealed gene expression changes related to oxidative phosphorylation, lysosome function, and lipid metabolism. Single-cell RNA sequencing revealed an increased proportion of DLAM clusters in cytokine mix-treated THP-1 macrophages. Functional assays demonstrated alterations in cell motility, phagocytosis, lysosomal activity, and metabolic and energetic profiles. Our findings provide insights into the cytokine-mediated reprogramming of macrophages towards disease-relevant states, highlighting their role in neurodegenerative diseases and potential for therapeutic development.
Collapse
Affiliation(s)
- Anna Podlesny-Drabiniok
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Carmen Romero-Molina
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Tulsi Patel
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Wen Yi See
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Yiyuan Liu
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Edoardo Marcora
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Alison M. Goate
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| |
Collapse
|
59
|
Kishishita S, Usui-Ouchi A, Ouchi Y, Hata Y, Ebihara N, Nakao S. Proliferative Diabetic Retinopathy Microenvironment Drives Microglial Polarization and Promotes Angiogenesis and Fibrosis via Cyclooxygenase-2/Prostaglandin E2 Signaling. Int J Mol Sci 2024; 25:11307. [PMID: 39457089 PMCID: PMC11508523 DOI: 10.3390/ijms252011307] [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: 09/14/2024] [Revised: 10/15/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Diabetic retinopathy (DR) is the leading cause of visual impairment, particularly in the proliferative form (proliferative DR [PDR]). The impact of the PDR microenvironment on microglia, which are the resident immune cells in the central nervous system, and the specific pathological changes it may induce remain unclear. This study aimed to investigate the role of microglia in the progression of PDR under hypoxic and inflammatory conditions. We performed a comprehensive gene expression analysis using human-induced pluripotent stem cell-derived microglia under different stimuli (dimethyloxalylglycine (DMOG), lipopolysaccharide (LPS), and DMOG + LPS) to mimic the hypoxic inflammatory environment characteristic of PDR. Principal component analysis revealed distinct gene expression profiles, with 76 genes synergistically upregulated under combined stimulation. Notably, prostaglandin-endoperoxide synthase 2 (encoding cyclooxygenase (COX)-2) exhibited the most pronounced increase, leading to elevated prostaglandin E2 (PGE2) levels and driving pathological angiogenesis and inflammation via the COX-2/PGE2/PGE receptor 2 signaling axis. Additionally, the upregulation of the fibrogenic genes snail family transcriptional repressor 1 and collagen type I alpha 1 chain suggested a role for microglia in fibrosis. These findings underscore the critical involvement of microglia in PDR and suggest that targeting both the angiogenic and fibrotic pathways may present new therapeutic strategies for managing this condition.
Collapse
Affiliation(s)
- Shuta Kishishita
- Department of Ophthalmology, Juntendo University Urayasu Hospital, 2-1-1 Urayasu, Chiba 279-0021, Japan
| | - Ayumi Usui-Ouchi
- Department of Ophthalmology, Juntendo University Urayasu Hospital, 2-1-1 Urayasu, Chiba 279-0021, Japan
| | - Yasuo Ouchi
- Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba 263-8522, Japan
| | - Yuiko Hata
- Department of Ophthalmology, Juntendo University Urayasu Hospital, 2-1-1 Urayasu, Chiba 279-0021, Japan
| | - Nobuyuki Ebihara
- Department of Ophthalmology, Juntendo University Urayasu Hospital, 2-1-1 Urayasu, Chiba 279-0021, Japan
| | - Shintaro Nakao
- Department of Ophthalmology, Juntendo University Graduate school of Medicine, 2-1-1 Bunkyo, Tokyo 113-8421, Japan
| |
Collapse
|
60
|
Warden AS, Salem NA, Brenner E, Sutherland GT, Stevens J, Kapoor M, Goate AM, Dayne Mayfield R. Integrative genomics approach identifies glial transcriptomic dysregulation and risk in the cortex of individuals with Alcohol Use Disorder. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.16.607185. [PMID: 39211266 PMCID: PMC11360965 DOI: 10.1101/2024.08.16.607185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Alcohol use disorder (AUD) is a prevalent neuropsychiatric disorder that is a major global health concern, affecting millions of people worldwide. Past molecular studies of AUD used underpowered single cell analysis or bulk homogenates of postmortem brain tissue, which obscures gene expression changes in specific cell types. Here we performed single nuclei RNA-sequencing analysis of 73 post-mortem samples from individuals with AUD (N=36, N nuclei = 248,873) and neurotypical controls (N=37, N nuclei = 210,573) in both sexes across two institutional sites. We identified 32 clusters and found widespread cell type-specific transcriptomic changes across the cortex in AUD, particularly affecting glia. We found the greatest dysregulation in novel microglial and astrocytic subtypes that accounted for the majority of differential gene expression and co-expression modules linked to AUD. Analysis for cell type-specific enrichment of aggregate genetic risk for AUD identified subtypes of microglia and astrocytes as potential key players not only affected by but causally linked to the progression of AUD. These results highlight the importance of cell-type specific molecular changes in AUD and offer opportunities to identify novel targets for treatment.
Collapse
|
61
|
Mirarchi A, Albi E, Arcuri C. Microglia Signatures: A Cause or Consequence of Microglia-Related Brain Disorders? Int J Mol Sci 2024; 25:10951. [PMID: 39456734 PMCID: PMC11507570 DOI: 10.3390/ijms252010951] [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: 08/09/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Microglia signatures refer to distinct gene expression profiles or patterns of gene activity that are characteristic of microglia. Advances in gene expression profiling techniques, such as single-cell RNA sequencing, have allowed us to study microglia at a more detailed level and identify unique gene expression patterns that are associated, but not always, with different functional states of these cells. Microglial signatures depend on the developmental stage, brain region, and specific pathological conditions. By studying these signatures, it has been possible to gain insights into the underlying mechanisms of microglial activation and begin to develop targeted therapies to modulate microglia-mediated immune responses in the CNS. Historically, the first two signatures coincide with M1 pro-inflammatory and M2 anti-inflammatory phenotypes. The first one includes upregulation of genes such as CD86, TNF-α, IL-1β, and iNOS, while the second one may involve genes like CD206, Arg1, Chil3, and TGF-β. However, it has long been known that many and more specific phenotypes exist between M1 and M2, likely with corresponding signatures. Here, we discuss specific microglial signatures and their association, if any, with neurodegenerative pathologies and other brain disorders.
Collapse
Affiliation(s)
- Alessandra Mirarchi
- Department of Medicine and Surgery, University of Perugia, Piazza L. Severi 1, 06132 Perugia, Italy;
| | - Elisabetta Albi
- Department of Pharmaceutical Sciences, University of Perugia, Via Fabretti 48, 06123 Perugia, Italy;
| | - Cataldo Arcuri
- Department of Medicine and Surgery, University of Perugia, Piazza L. Severi 1, 06132 Perugia, Italy;
| |
Collapse
|
62
|
Yang Z, Zhang H, Yuan Z, Chen J, Zheng G, Zou S. The effects of GCRV on various tissues of grass carp (Ctenopharyngodon idella) and identification of differential interferon-stimulating genes (ISGs) through muscle transcriptome analysis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:116956. [PMID: 39208574 DOI: 10.1016/j.ecoenv.2024.116956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/24/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
Grass carp hemorrhagic disease is caused by the grass carp reovirus (GCRV). The disease spreads rapidly and has a high fatality rate, which seriously affects grass carp culture. Moreover, the molecular mechanisms underlying grass carp hemorrhagic disease remain unclear. To decipher the effects of GCRV on grass carp tissues, resistant grass carp A (GA) and susceptible grass carp B (GB) were selected through GCRV treatment, and control grass carp C (GC) was also established. The gill, liver, and muscle tissues exhibited different onset symptoms under the influence of GCRV by histological observation. We selected muscle samples with significant differences in symptoms for Illumina RNA sequencing. Analyses using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes revealed 3512, 3074, and 1853 differentially expressed genes between "GC vs. GB," "GC vs. GA," and "GA vs. GB," respectively. Additionally, 40 differential immune-related genes and 28 differential interferon-stimulating genes (ISGs) related to the interferon (IFN) pathway were identified. The expression of immunogene-related genes of GB and GA, such as MDA5, IL-34, NF-KB, TRIM25, SOCS3, CEBPB, and BCL2, and genes associated with the JAK-STAT signaling pathway, such as IRF4, STAT1, STAT3, JAK 1, and JAK 2, was significantly upregulated. The IFN and JAK-STAT signaling pathways were closely related to anti-GCRV infection. The transcriptome data and predicted immune genes and ISGs in this study provide novel insights into the treatment of GCRV.
Collapse
Affiliation(s)
- Ziquan Yang
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China; Genetics and Breeding Center for Blunt Snout Bream, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China
| | - Huimei Zhang
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China; Genetics and Breeding Center for Blunt Snout Bream, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China
| | - Ziming Yuan
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China; Genetics and Breeding Center for Blunt Snout Bream, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China
| | - Jie Chen
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China; Genetics and Breeding Center for Blunt Snout Bream, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China
| | - Guodong Zheng
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China; Genetics and Breeding Center for Blunt Snout Bream, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China.
| | - Shuming Zou
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China; Genetics and Breeding Center for Blunt Snout Bream, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China.
| |
Collapse
|
63
|
Hofer MJ, Modesti N, Coufal NG, Wang Q, Sase S, Miner J, Vanderver A, Bennett ML. The prototypical interferonopathy: Aicardi-Goutières syndrome from bedside to bench. Immunol Rev 2024; 327:83-99. [PMID: 39473130 PMCID: PMC11672868 DOI: 10.1111/imr.13413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2024]
Abstract
Aicardi-Goutières syndrome (AGS) is a progressive genetic encephalopathy caused by pathogenic mutations in genes controlling cellular anti-viral responses and nucleic acid metabolism. The mutations initiate autoinflammatory processes in the brain and systemically that are triggered by chronic overproduction of type I interferon (IFN), including IFN-alpha. Emerging disease-directed therapies aim to dampen autoinflammation and block cellular responses to IFN production, creating an urgent and unmet need to understand better which cells, compartments, and mechanisms underlying disease pathogenesis. In this review, we highlight existing pre-clinical models of AGS and our current understanding of how causative genetic mutations promote disease in AGS, to promote new model development and a continued focus on improving and directing future therapies.
Collapse
Affiliation(s)
- Markus J. Hofer
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, NSW 2006, Australia; NHMRC Ideas Grant to MJH APP2001543
| | - Nicholson Modesti
- Division of Neurology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104
| | - Nicole G. Coufal
- Department of Pediatrics, University of California, San Diego CA 92093, Rady Children’s Hospital, San Diego CA 92123. Sanford Consortium for Regenerative Medicine, San Diego CA 92037
| | - Qingde Wang
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213
| | - Sunetra Sase
- Division of Neurology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104
| | - Jonathan Miner
- Departments of Medicine and Microbiology, RVCL Research Center, and Colton Center for Autoimmunity, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104
| | - Adeline Vanderver
- Division of Neurology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104
| | - Mariko L Bennett
- Division of Neurology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104
| |
Collapse
|
64
|
Carrillo GL, Su J, Cawley ML, Wei D, Gill SK, Blader IJ, Fox MA. Complement-dependent loss of inhibitory synapses on pyramidal neurons following Toxoplasma gondii infection. J Neurochem 2024; 168:3365-3385. [PMID: 36683435 PMCID: PMC10363253 DOI: 10.1111/jnc.15770] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 01/06/2023] [Accepted: 01/15/2023] [Indexed: 01/24/2023]
Abstract
The apicomplexan parasite Toxoplasma gondii has developed mechanisms to establish a central nervous system infection in virtually all warm-blooded animals. Acute T. gondii infection can cause neuroinflammation, encephalitis, and seizures. Meanwhile, studies in humans, nonhuman primates, and rodents have linked chronic T. gondii infection with altered behavior and increased risk for neuropsychiatric disorders, including schizophrenia. These observations and associations raise questions about how this parasitic infection may alter neural circuits. We previously demonstrated that T. gondii infection triggers the loss of inhibitory perisomatic synapses, a type of synapse whose dysfunction or loss has been linked to neurological and neuropsychiatric disorders. We showed that phagocytic cells (including microglia and infiltrating monocytes) contribute to the loss of these inhibitory synapses. Here, we show that these phagocytic cells specifically ensheath excitatory pyramidal neurons, leading to the preferential loss of perisomatic synapses on these neurons and not those on cortical interneurons. Moreover, we show that infection induces an increased expression of the complement C3 gene, including by populations of these excitatory neurons. Infecting C3-deficient mice with T. gondii revealed that C3 is required for the loss of perisomatic inhibitory synapses. Interestingly, loss of C1q did not prevent the loss of perisomatic synapses following infection. Together, these findings provide evidence that T. gondii induces changes in excitatory pyramidal neurons that trigger the selective removal of inhibitory perisomatic synapses and provide a role for a nonclassical complement pathway in the remodeling of inhibitory circuits in the infected brain.
Collapse
Affiliation(s)
- Gabriela L. Carrillo
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, 24016, USA
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, Virginia, 24061, USA
| | - Jianmin Su
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, 24016, USA
- School of Neuroscience, College of Science, Virginia Tech, Blacksburg, Virginia, 24061, USA
| | - Mikel L. Cawley
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, 24016, USA
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, Virginia, 24061, USA
| | - Derek Wei
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, 24016, USA
- School of Neuroscience, College of Science, Virginia Tech, Blacksburg, Virginia, 24061, USA
| | - Simran K. Gill
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, 24016, USA
- Department of Psychology, Roanoke College, Salem, Virginia, 24153, USA
- NeuroSURF Program, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, 24016, USA
| | - Ira J. Blader
- Department of Microbiology and Immunology, University at Buffalo, Buffalo, New York, 14203, USA
| | - Michael A. Fox
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, 24016, USA
- School of Neuroscience, College of Science, Virginia Tech, Blacksburg, Virginia, 24061, USA
- Department of Biological Sciences, College of Science, Virginia Tech, Blacksburg, Virginia, 24061, USA
- Department of Pediatrics, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, 24016, USA
| |
Collapse
|
65
|
Gao ML, Wang TY, Lin X, Tang C, Li M, Bai ZP, Liu ZC, Chen LJ, Kong QR, Pan SH, Zeng SS, Guo Y, Cai JQ, Huang XF, Zhang J. Retinal Organoid Microenvironment Enhanced Bioactivities of Microglia-Like Cells Derived From HiPSCs. Invest Ophthalmol Vis Sci 2024; 65:19. [PMID: 39392440 PMCID: PMC11472886 DOI: 10.1167/iovs.65.12.19] [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: 04/27/2024] [Accepted: 07/12/2024] [Indexed: 10/12/2024] Open
Abstract
Purpose Microglia-like cells derived from stem cells (iMG) provide a plentiful cell source for studying the functions of microglia in both normal and pathological conditions. Our goal is to establish a simplified and effective method for generating iMG in a precisely defined system. Additionally, we aim to achieve functional maturation of iMG through coculture with retinal organoids. Methods In this study, iMG were produced under precisely defined conditions. They were subjected to LPS and poly IC stimulation. Additionally, we examined distinct phenotypic and functional variances between iMG and HMC3, a commonly used human microglia cell line. To investigate how the retinal cell interaction enhances microglial properties, iMG were cocultured with retinal organoids, producing CC-iMG. We performed RNA sequencing, electrophysiological analysis, and transmission electron microscope (TEM) to examine the maturation of CC-iMG compared to iMG. Results Our results demonstrated that iMG performed immune-responsive profiles closely resembling those of primary human microglia. Compared to HMC3, iMG expressed a higher level of typical microglial markers and exhibited enhanced phagocytic activity. The transcriptomic analysis uncovered notable alterations in the ion channel profile of CC-iMG compared to iMG. Electrophysiological examination demonstrated a heightened intensity of inward- and outward-rectifying K+ currents in CC-iMG. Furthermore, CC-iMG displayed elevated numbers of lysosomes and mitochondria, coupled with increased phagocytic activity. Conclusions These findings contribute to advancing our understanding of human microglial biology, specifically in characterizing and elucidating the functions of CC-iMG, thereby offering an in vitro microglial model for future scientific research and potential clinical applications in cell therapy.
Collapse
Affiliation(s)
- Mei-Ling Gao
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Tong-Yu Wang
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Xin Lin
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Chun Tang
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Mengyao Li
- Department of Critical Care Medicine, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 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, China
| | - Zhi-Cong Liu
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Li-Jun Chen
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Qing-Ran Kong
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shao-Hui Pan
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, China
| | - Shan-Shan Zeng
- China National Institute of Standardization, Beijing, China
| | - Ya Guo
- China National Institute of Standardization, Beijing, China
| | - Jian-Qi Cai
- China National Institute of Standardization, Beijing, 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, China
| | - Jun Zhang
- The State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, China
- Laboratory of Retinal Physiology and Disease, Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
66
|
Schlachetzki JC, Gianella S, Ouyang Z, Lana AJ, Yang X, O'Brien S, Challacombe JF, Gaskill PJ, Jordan-Sciutto KL, Chaillon A, Moore D, Achim CL, Ellis RJ, Smith DM, Glass CK. Gene expression and chromatin conformation of microglia in virally suppressed people with HIV. Life Sci Alliance 2024; 7:e202402736. [PMID: 39060113 PMCID: PMC11282357 DOI: 10.26508/lsa.202402736] [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: 03/25/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
The presence of HIV in sequestered reservoirs is a central impediment to a functional cure, allowing HIV to persist despite life-long antiretroviral therapy (ART), and driving a variety of comorbid conditions. Our understanding of the latent HIV reservoir in the central nervous system is incomplete, because of difficulties in accessing human central nervous system tissues. Microglia contribute to HIV reservoirs, but the molecular phenotype of HIV-infected microglia is poorly understood. We leveraged the unique "Last Gift" rapid autopsy program, in which people with HIV are closely followed until days or even hours before death. Microglial populations were heterogeneous regarding their gene expression profiles but showed similar chromatin accessibility landscapes. Despite ART, we detected occasional microglia containing cell-associated HIV RNA and HIV DNA integrated into open regions of the host's genome (∼0.005%). Microglia with detectable HIV RNA showed an inflammatory phenotype. These results demonstrate a distinct myeloid cell reservoir in the brains of people with HIV despite suppressive ART. Strategies for curing HIV and neurocognitive impairment will need to consider the myeloid compartment to be successful.
Collapse
Affiliation(s)
- Johannes Cm Schlachetzki
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, CA, USA
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA
| | - Sara Gianella
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, San Diego, CA, USA
| | - Zhengyu Ouyang
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, CA, USA
| | - Addison J Lana
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, CA, USA
| | - Xiaoxu Yang
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Sydney O'Brien
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, CA, USA
| | - Jean F Challacombe
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, CA, USA
| | - Peter J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Kelly L Jordan-Sciutto
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Antoine Chaillon
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, San Diego, CA, USA
| | - David Moore
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Cristian L Achim
- Department of Pathology, University of California San Diego, San Diego, CA, USA
| | - Ronald J Ellis
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA
| | - Davey M Smith
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, San Diego, CA, USA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
67
|
Kim J, Sullivan O, Lee K, Jao J, Tamayo J, Madany AM, Wong B, Ashwood P, Ciernia AV. Repeated LPS induces training and tolerance of microglial responses across brain regions. J Neuroinflammation 2024; 21:233. [PMID: 39304952 PMCID: PMC11414187 DOI: 10.1186/s12974-024-03198-1] [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: 04/08/2024] [Accepted: 08/08/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Neuroinflammation is involved in the pathogenesis of almost every central nervous system disorder. As the brain's innate immune cells, microglia fine tune their activity to a dynamic brain environment. Previous studies have shown that repeated bouts of peripheral inflammation can trigger long-term changes in microglial gene expression and function, a form of innate immune memory. METHODS AND RESULTS In this study, we used multiple low-dose lipopolysaccharide (LPS) injections in adult mice to study the acute cytokine, transcriptomic, and microglia morphological changes that contribute to the formation of immune memory in the frontal cortex, hippocampus, and striatum, as well as the long-term effects of these changes on behavior. Training and tolerance of gene expression was shared across regions, and we identified 3 unique clusters of DEGs (2xLPS-sensitive, 4xLPS-sensitive, LPS-decreased) enriched for different biological functions. 2xLPS-sensitive DEG promoters were enriched for binding sites for IRF and NFkB family transcription factors, two key regulators of innate immune memory. We quantified shifts in microglia morphological populations and found that while the proportion of ramified and rod-like microglia mostly remained consistent within brain regions and sexes with LPS treatment, there was a shift from ameboid towards hypertrophic morphological states across immune memory states and a dynamic emergence and resolution of events of microglia aligning end-to-end with repeated LPS. CONCLUSIONS Together, findings support the dynamic regulation of microglia during the formation of immune memories in the brain and support future work to exploit this model in brain disease contexts.
Collapse
Affiliation(s)
- Jennifer Kim
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Olivia Sullivan
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Kristen Lee
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Justin Jao
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Juan Tamayo
- MIND Institute, University of California Davis, Davis, USA
| | | | - Brandon Wong
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Paul Ashwood
- MIND Institute, University of California Davis, Davis, USA
| | - Annie Vogel Ciernia
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada.
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada.
| |
Collapse
|
68
|
Nemec KM, Uy G, Chaluvadi VS, Purnell FS, Elfayoumi B, O'Brien CA, Aisenberg WH, Lombroso SI, Guo X, Blank N, Oon CH, Yaqoob F, Temsamrit B, Rawat P, Thaiss CA, Wang Q, Bennett ML, Bennett FC. Microglia replacement by ER-Hoxb8 conditionally immortalized macrophages provides insight into Aicardi-Goutières Syndrome neuropathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.613629. [PMID: 39345609 PMCID: PMC11430044 DOI: 10.1101/2024.09.18.613629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Microglia, the brain's resident macrophages, can be reconstituted by surrogate cells - a process termed "microglia replacement." To expand the microglia replacement toolkit, we here introduce estrogen-regulated (ER) homeobox B8 (Hoxb8) conditionally immortalized macrophages, a cell model for generation of immune cells from murine bone marrow, as a versatile model for microglia replacement. We find that ER-Hoxb8 macrophages are highly comparable to primary bone marrow-derived (BMD) macrophages in vitro, and, when transplanted into a microglia-free brain, engraft the parenchyma and differentiate into microglia-like cells. Furthermore, ER-Hoxb8 progenitors are readily transducible by virus and easily stored as stable, genetically manipulated cell lines. As a demonstration of this system's power for studying the effects of disease mutations on microglia in vivo, we created stable, Adar1-mutated ER-Hoxb8 lines using CRISPR-Cas9 to study the intrinsic contribution of macrophages to Aicardi-Goutières Syndrome (AGS), an inherited interferonopathy that primarily affects the brain and immune system. We find that Adar1 knockout elicited interferon secretion and impaired macrophage production in vitro, while preventing brain macrophage engraftment in vivo - phenotypes that can be rescued with concurrent mutation of Ifih1 (MDA5) in vitro, but not in vivo. Lastly, we extended these findings by generating ER-Hoxb8 progenitors from mice harboring a patient-specific Adar1 mutation (D1113H). We demonstrated the ability of microglia-specific D1113H mutation to drive interferon production in vivo, suggesting microglia drive AGS neuropathology. In sum, we introduce the ER-Hoxb8 approach to model microglia replacement and use it to clarify macrophage contributions to AGS.
Collapse
Affiliation(s)
- Kelsey M Nemec
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Genevieve Uy
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - V Sai Chaluvadi
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Freddy S Purnell
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Epigenetics Institute, University of Pennsylvania, Perelman School of Medicine. Philadelphia, PA, USA
| | - Bilal Elfayoumi
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carleigh A O'Brien
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - William H Aisenberg
- 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
| | - Xinfeng Guo
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Niklas Blank
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Chet Huan Oon
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fazeela Yaqoob
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Brian Temsamrit
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Priyanka Rawat
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Christoph A Thaiss
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Qingde Wang
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mariko L Bennett
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 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
| |
Collapse
|
69
|
Petry P, Aktories P, Oschwald A, Kierdorf K. Protocol for long-term monocultures of murine macrophages derived from distinct adult tissues. STAR Protoc 2024; 5:103145. [PMID: 38909360 PMCID: PMC11245975 DOI: 10.1016/j.xpro.2024.103145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/29/2024] [Accepted: 06/03/2024] [Indexed: 06/25/2024] Open
Abstract
Tissue-resident macrophages (TRMs) constitute the first line of defense against infection in all organs and perform organ-specific functions during tissue homeostasis. Here, we present a protocol for long-term monocultures of murine macrophages from different adult organs, including the brain, liver, peritoneal cavity, and lung. We describe steps for tissue preparation and the use of a combination of organotypic conditions to maintain a TRM-like identity in vitro, resulting in an ideal screening platform for a wide range of assays and readouts. For complete details on the use and execution of this protocol, please refer to Aktories et al.1.
Collapse
Affiliation(s)
- Philippe Petry
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Philipp Aktories
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany.
| | - Alexander Oschwald
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Katrin Kierdorf
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; CIBSS-Center for Integrative Biological Signaling Studies, University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
70
|
Guvenek A, Parikshak N, Zamolodchikov D, Gelfman S, Moscati A, Dobbyn L, Stahl E, Shuldiner A, Coppola G. Transcriptional profiling in microglia across physiological and pathological states identifies a transcriptional module associated with neurodegeneration. Commun Biol 2024; 7:1168. [PMID: 39294270 PMCID: PMC11411103 DOI: 10.1038/s42003-024-06684-7] [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: 08/08/2023] [Accepted: 08/06/2024] [Indexed: 09/20/2024] Open
Abstract
Microglia are the resident immune cells of the central nervous system and are involved in brain development, homeostasis, and disease. New imaging and genomics technologies are revealing microglial complexity across developmental and functional states, brain regions, and diseases. We curated a set of publicly available gene expression datasets from human microglia spanning disease and health to identify sets of genes reflecting physiological and pathological microglial states. We also integrated multiple human microglial single-cell RNA-seq datasets in Alzheimer's disease (AD), multiple sclerosis (MS), and Parkinson's disease, and identified a distinct microglial transcriptional signature shared across diseases. Analysis of germ-line DNA identified genes with variants associated with AD and MS that are overrepresented in microglial gene sets, including the disease-associated transcriptional signature. This work points to genes that are dysregulated in disease states and provides a resource for the analysis of diseases in which microglia are implicated by genetic evidence.
Collapse
Affiliation(s)
- Aysegul Guvenek
- Regeneron Genetics Center, Tarrytown, NY, USA
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | | | | | | | | | - Lee Dobbyn
- Regeneron Genetics Center, Tarrytown, NY, USA
| | - Eli Stahl
- Regeneron Genetics Center, Tarrytown, NY, USA
| | | | | |
Collapse
|
71
|
Yadav H, Bakshi A, Anamika, Singh V, Paul P, Murugan NA, Maurya SK. Co-localization and co-expression of Olfml3 with Iba1 in brain of mice. J Neuroimmunol 2024; 394:578411. [PMID: 39079458 DOI: 10.1016/j.jneuroim.2024.578411] [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: 06/11/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/30/2024]
Abstract
Olfml3 is a microglia-specific protein whose role in neuroinflammation is elusive. In silico analysis was conducted to characterize the Olfml3 protein, followed by molecular docking and MD simulation to check possible interaction with Iba1. Further, expression and co-localization analysis was performed in the LPS-induced neuroinflammatory mice brains. Results suggest that Olfml3 physically interacts with Iba1. Olfml3 and Iba1 expression increases during neuroinflammation in mice brains. Olfml3 was observed to co-localize with Iba1, and the number of Olfml3 and Iba1 dual-positive cells increased in the brain of the neuroinflammatory mice model. Thus, Olfml3 could potentially participate in microglia functions by interacting with Iba1.
Collapse
Affiliation(s)
- Himanshi Yadav
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Amrita Bakshi
- Department of Zoology, Ramjas College, University of Delhi, Delhi, India
| | - Anamika
- Department of Zoology, Ramjas College, University of Delhi, Delhi, India
| | - Vishal Singh
- Electron Microscope Facility, All India Institute of Medical Sciences, New Delhi, India
| | - Prateek Paul
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Industrial Estate, Delhi, India
| | - N Arul Murugan
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Industrial Estate, Delhi, India
| | - Shashank Kumar Maurya
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India.
| |
Collapse
|
72
|
Liu J, Lei F, Yan B, Cui N, Sharma J, Correa V, Roach L, Nicolaou S, Pitts K, Chodosh J, Maidana DE, Vavvas D, Margeta MA, Zhang H, Weitz D, Mostoslavsky R, Paschalis EI. Epigenetic adaptation drives monocyte differentiation into microglia-like cells upon engraftment into the retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.09.612126. [PMID: 39314467 PMCID: PMC11419019 DOI: 10.1101/2024.09.09.612126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The identification of specific markers for microglia has been a long-standing challenge. Recently, markers such as P2ry12, TMEM119, and Fcrls have been proposed as microglia-specific and widely used to explore microglial functions within various central nervous system (CNS) contexts. The specificity of these markers was based on the assumption that circulating monocytes retain their distinct signatures even after infiltrating the CNS. However, recent findings reveal that infiltrating monocytes can adopt microglia-like characteristics while maintaining a pro-inflammatory profile upon permanent engraftment in the CNS.In this study, we utilize bone marrow chimeras, single-cell RNA sequencing, ATAC-seq, flow cytometry, and immunohistochemistry to demonstrate that engrafted monocytes acquire expression of established microglia markers-P2ry12, TMEM119, Fcrls-and the pan-myeloid marker Iba1, which has been commonly mischaracterized as microglia-specific. These changes are accompanied by alterations in chromatin accessibility and shifts in chromatin binding motifs that are indicative of microglial identity. Moreover, we show that engrafted monocytes dynamically regulate the expression of CX3CR1, CCR2, Ly6C, and transcription factors PU.1, CTCF, RUNX, AP-1, CEBP, and IRF2, all of which are crucial for shaping microglial identity. This study is the first to illustrate that engrafted monocytes in the retina undergo both epigenetic and transcriptional changes, enabling them to express microglia-like signatures. These findings highlight the need for future research to account for these changes when assessing the roles of monocytes and microglia in CNS pathology.
Collapse
Affiliation(s)
- Jie Liu
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Fengyang Lei
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Bin Yan
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Naiwen Cui
- School of Engineering and Applied Sciences (SEAS), Harvard University, Cambridge, MA, USA
| | - Jyoti Sharma
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Victor Correa
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Lara Roach
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Savvas Nicolaou
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Kristen Pitts
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - James Chodosh
- Department of Ophthalmology and Visual Sciences, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Daniel E. Maidana
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL
| | - Demetrios Vavvas
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Milica A Margeta
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Huidan Zhang
- School of Engineering and Applied Sciences (SEAS), Harvard University, Cambridge, MA, USA
| | - David Weitz
- School of Engineering and Applied Sciences (SEAS), Harvard University, Cambridge, MA, USA
| | - Raul Mostoslavsky
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Eleftherios I. Paschalis
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
73
|
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
|
74
|
Wogram E, Sümpelmann F, Dong W, Rawat E, Fernández Maestre I, Fu D, Braswell B, Khalil A, Buescher JM, Mittler G, Borner GHH, Vlachos A, Tholen S, Schilling O, Bell GW, Rambold AS, Akhtar A, Schnell O, Beck J, Abu-Remaileh M, Prinz M, Jaenisch R. Rapid phagosome isolation enables unbiased multiomic analysis of human microglial phagosomes. Immunity 2024; 57:2216-2231.e11. [PMID: 39151426 DOI: 10.1016/j.immuni.2024.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 01/25/2024] [Accepted: 07/19/2024] [Indexed: 08/19/2024]
Abstract
Microglia are the resident macrophages of the central nervous system (CNS). Their phagocytic activity is central during brain development and homeostasis-and in a plethora of brain pathologies. However, little is known about the composition, dynamics, and function of human microglial phagosomes under homeostatic and pathological conditions. Here, we developed a method for rapid isolation of pure and intact phagosomes from human pluripotent stem cell-derived microglia under various in vitro conditions, and from human brain biopsies, for unbiased multiomic analysis. Phagosome profiling revealed that microglial phagosomes were equipped to sense minute changes in their environment and were highly dynamic. We detected proteins involved in synapse homeostasis, or implicated in brain pathologies, and identified the phagosome as the site where quinolinic acid was stored and metabolized for de novo nicotinamide adenine dinucleotide (NAD+) generation in the cytoplasm. Our findings highlight the central role of phagosomes in microglial functioning in the healthy and diseased brain.
Collapse
Affiliation(s)
- Emile Wogram
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Felix Sümpelmann
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Wentao Dong
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA; The Institute for Chemistry, Engineering & Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
| | - Eshaan Rawat
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA; The Institute for Chemistry, Engineering & Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
| | | | - Dongdong Fu
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Brandyn Braswell
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Andrew Khalil
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; The Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA; Harvard John A. Paulson School of Engineering and Applied Sciences, Boston, MA 02134, USA
| | - Joerg M Buescher
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Gerhard Mittler
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Georg H H Borner
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Stefan Tholen
- Institute of Surgical Pathology, Medical Center, University of Freiburg, 79106 Freiburg, Germany
| | - Oliver Schilling
- Institute of Surgical Pathology, Medical Center, University of Freiburg, 79106 Freiburg, Germany
| | - George W Bell
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Angelika S Rambold
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Asifa Akhtar
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Oliver Schnell
- Department of Neurosurgery, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Jürgen Beck
- Department of Neurosurgery, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - 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 & Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; BIOSS Centre for Biological Signalling Studies and Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, 79104 Freiburg, Germany
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| |
Collapse
|
75
|
Wei B, Cheng G, Bi Q, Lu C, Sun Q, Li L, Chen N, Hu M, Lu H, Xu X, Mao G, Wan S, Hu Z, Gu Y, Zheng J, Zhao L, Shen XZ, Liu X, Shi P. Microglia in the hypothalamic paraventricular nucleus sense hemodynamic disturbance and promote sympathetic excitation in hypertension. Immunity 2024; 57:2030-2042.e8. [PMID: 39116878 DOI: 10.1016/j.immuni.2024.07.011] [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: 09/06/2023] [Revised: 04/22/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024]
Abstract
Hypertension is usually accompanied by elevated sympathetic tonicity, but how sympathetic hyperactivity is triggered is not clear. Recent advances revealed that microglia-centered neuroinflammation contributes to sympathetic excitation in hypertension. In this study, we performed a temporospatial analysis of microglia at both morphological and transcriptomic levels and found that microglia in the hypothalamic paraventricular nucleus (PVN), a sympathetic center, were early responders to hypertensive challenges. Vasculature analyses revealed that the PVN was characterized by high capillary density, thin vessel diameter, and complex vascular topology relative to other brain regions. As such, the PVN was susceptible to the penetration of ATP released from the vasculature in response to hemodynamic disturbance after blood pressure increase. Mechanistically, ATP ligation to microglial P2Y12 receptor was responsible for microglial inflammatory activation and the eventual sympathetic overflow. Together, these findings identified a distinct vasculature pattern rendering vulnerability of PVN pre-sympathetic neurons to hypertension-associated microglia-mediated inflammatory insults.
Collapse
Affiliation(s)
- Bo Wei
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Guo Cheng
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Qianqian Bi
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Cheng Lu
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Qihang Sun
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Li Li
- Department of Pharmacy, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, China
| | - Ningting Chen
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Miner Hu
- Department of Cardiology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310013, China
| | - Haoran Lu
- Zhejiang University, University of Edinburgh Institute, Zhejiang University School of Medicine, Haining 314400, China
| | - Xuancheng Xu
- Zhejiang Chinese Medical University, Hangzhou 310013, China; Department of Neurology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, China
| | - Genxiang Mao
- Zhejiang Provincial Key Lab of Geriatrics, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Shu Wan
- Brain Center, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, China
| | - Zhechun Hu
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, Zhejiang University, Hangzhou 310058, China; Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Gu
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, Zhejiang University, Hangzhou 310058, China; Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaxin Zheng
- Key Laboratory for Biomedical Engineering of Ministrey of Education, Collage of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310013, China
| | - Li Zhao
- Key Laboratory for Biomedical Engineering of Ministrey of Education, Collage of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310013, China
| | - Xiao Z Shen
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Brain Center, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, China; Department of Physiology, Zhejiang University School of Medicine, Hangzhou 310058, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310013, China
| | - Xiaoli Liu
- Department of Neurology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, China.
| | - Peng Shi
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310013, China.
| |
Collapse
|
76
|
Guzmán-Ruíz MA, Guerrero Vargas NN, Ramírez-Carreto RJ, González-Orozco JC, Torres-Hernández BA, Valle-Rodríguez M, Guevara-Guzmán R, Chavarría A. Microglia in physiological conditions and the importance of understanding their homeostatic functions in the arcuate nucleus. Front Immunol 2024; 15:1392077. [PMID: 39295865 PMCID: PMC11408222 DOI: 10.3389/fimmu.2024.1392077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 08/20/2024] [Indexed: 09/21/2024] Open
Abstract
Microglia are highly dynamic cells that have been mainly studied under pathological conditions. The present review discusses the possible implication of microglia as modulators of neuronal electrical responses in physiological conditions and hypothesizes how these cells might modulate hypothalamic circuits in health and during obesity. Microglial cells studied under physiological conditions are highly diverse, depending on the developmental stage and brain region. The evidence also suggests that neuronal electrical activity modulates microglial motility to control neuronal excitability. Additionally, we show that the expression of genes associated with neuron-microglia interaction is down-regulated in obese mice compared to control-fed mice, suggesting an alteration in the contact-dependent mechanisms that sustain hypothalamic arcuate-median eminence neuronal function. We also discuss the possible implication of microglial-derived signals for the excitability of hypothalamic neurons during homeostasis and obesity. This review emphasizes the importance of studying the physiological interplay between microglia and neurons to maintain proper neuronal circuit function. It aims to elucidate how disruptions in the normal activities of microglia can adversely affect neuronal health.
Collapse
Affiliation(s)
- Mara A Guzmán-Ruíz
- Programa de Becas Post-doctorales, Dirección General de Asuntos del Personal Académico, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Natalí N Guerrero Vargas
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Ricardo Jair Ramírez-Carreto
- Unidad de Medicina Experimental "Ruy Pérez Tamayo", Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | | | - Michelle Valle-Rodríguez
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Rosalinda Guevara-Guzmán
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Anahí Chavarría
- Unidad de Medicina Experimental "Ruy Pérez Tamayo", Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
77
|
Koskuvi M, Pörsti E, Hewitt T, Räsänen N, Wu YC, Trontti K, McQuade A, Kalyanaraman S, Ojansuu I, Vaurio O, Cannon TD, Lönnqvist J, Therman S, Suvisaari J, Kaprio J, Blurton-Jones M, Hovatta I, Lähteenvuo M, Rolova T, Lehtonen Š, Tiihonen J, Koistinaho J. Genetic contribution to microglial activation in schizophrenia. Mol Psychiatry 2024; 29:2622-2633. [PMID: 38519640 PMCID: PMC11420079 DOI: 10.1038/s41380-024-02529-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 03/06/2024] [Accepted: 03/13/2024] [Indexed: 03/25/2024]
Abstract
Several lines of evidence indicate the involvement of neuroinflammatory processes in the pathophysiology of schizophrenia (SCZ). Microglia are brain resident immune cells responding toward invading pathogens and injury-related products, and additionally, have a critical role in improving neurogenesis and synaptic functions. Aberrant activation of microglia in SCZ is one of the leading hypotheses for disease pathogenesis, but due to the lack of proper human cell models, the role of microglia in SCZ is not well studied. We used monozygotic twins discordant for SCZ and healthy individuals to generate human induced pluripotent stem cell-derived microglia to assess the transcriptional and functional differences in microglia between healthy controls, affected twins and unaffected twins. The microglia from affected twins had increased expression of several common inflammation-related genes compared to healthy individuals. Microglia from affected twins had also reduced response to interleukin 1 beta (IL1β) treatment, but no significant differences in migration or phagocytotic activity. Ingenuity Pathway Analysis (IPA) showed abnormalities related to extracellular matrix signaling. RNA sequencing predicted downregulation of extracellular matrix structure constituent Gene Ontology (GO) terms and hepatic fibrosis pathway activation that were shared by microglia of both affected and unaffected twins, but the upregulation of major histocompatibility complex (MHC) class II receptors was observed only in affected twin microglia. Also, the microglia of affected twins had heterogeneous response to clozapine, minocycline, and sulforaphane treatments. Overall, despite the increased expression of inflammatory genes, we observed no clear functional signs of hyperactivation in microglia from patients with SCZ. We conclude that microglia of the patients with SCZ have gene expression aberrations related to inflammation response and extracellular matrix without contributing to increased microglial activation.
Collapse
Affiliation(s)
- Marja Koskuvi
- Neuroscience Center, University of Helsinki, Helsinki, Finland
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Elina Pörsti
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Tristen Hewitt
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Noora Räsänen
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Ying-Chieh Wu
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Kalevi Trontti
- Neuroscience Center, University of Helsinki, Helsinki, Finland
- SleepWell Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| | - Amanda McQuade
- Department of Neurobiology & Behavior, UC Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, CA, USA
| | | | - Ilkka Ojansuu
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland
| | - Olli Vaurio
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland
| | - Tyrone D Cannon
- Department of Psychology and Psychiatry, Yale University, New Haven, CT, USA
| | - Jouko Lönnqvist
- Mental Health Unit, Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki, Finland
- Department of Psychiatry, University of Helsinki, Helsinki, Finland
| | - Sebastian Therman
- Mental Health Unit, Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki, Finland
| | - Jaana Suvisaari
- Mental Health Unit, Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki, Finland
| | - Jaakko Kaprio
- Institute for Molecular Medicine FIMM, University of Helsinki, Helsinki, Finland
| | - Mathew Blurton-Jones
- Department of Neurobiology & Behavior, UC Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, CA, USA
| | - Iiris Hovatta
- Neuroscience Center, University of Helsinki, Helsinki, Finland
- SleepWell Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| | - Markku Lähteenvuo
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland
| | - Taisia Rolova
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Šárka Lehtonen
- Neuroscience Center, University of Helsinki, Helsinki, Finland
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jari Tiihonen
- Neuroscience Center, University of Helsinki, Helsinki, Finland
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jari Koistinaho
- Neuroscience Center, University of Helsinki, Helsinki, Finland.
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
78
|
Kuhn MK, Proctor EA. Microglial Drivers of Alzheimer's Disease Pathology: An Evolution of Diverse Participating States. Proteins 2024:10.1002/prot.26723. [PMID: 39219300 PMCID: PMC11871049 DOI: 10.1002/prot.26723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/05/2024] [Accepted: 06/12/2024] [Indexed: 09/04/2024]
Abstract
Microglia, the resident immune-competent cells of the brain, become dysfunctional in Alzheimer's disease (AD), and their aberrant immune responses contribute to the accumulation of pathological proteins and neuronal injury. Genetic studies implicate microglia in the development of AD, prompting interest in developing immunomodulatory therapies to prevent or ameliorate disease. However, microglia take on diverse functional states in disease, playing both protective and detrimental roles in AD, which largely overlap and may shift over the disease course, complicating the identification of effective therapeutic targets. Extensive evidence gathered using transgenic mouse models supports an active role of microglia in pathology progression, though results vary and can be contradictory between different types of models and the degree of pathology at the time of study. Here, we review microglial immune signaling and responses that contribute to the accumulation and spread of pathological proteins or directly affect neuronal health. We additionally explore the use of induced pluripotent stem cell (iPSC)-derived models to study living human microglia and how they have contributed to our knowledge of AD and may begin to fill in the gaps left by mouse models. Ultimately, mouse and iPSC-derived models have their own limitations, and a comprehensive understanding of microglial dysfunction in AD will only be established by an integrated view across models and an appreciation for their complementary viewpoints and limitations.
Collapse
Affiliation(s)
- Madison K. Kuhn
- Department of Biomedical Engineering, Penn State University
- Department of Neurosurgery, Penn State College of Medicine
- Department of Pharmacology, Penn State College of Medicine
- Center for Neural Engineering, Penn State University
| | - Elizabeth A. Proctor
- Department of Biomedical Engineering, Penn State University
- Department of Neurosurgery, Penn State College of Medicine
- Department of Pharmacology, Penn State College of Medicine
- Center for Neural Engineering, Penn State University
- Department of Engineering Science & Mechanics, Penn State University
| |
Collapse
|
79
|
Ji Y, McLean JL, Xu R. Emerging Human Pluripotent Stem Cell-Based Human-Animal Brain Chimeras for Advancing Disease Modeling and Cell Therapy for Neurological Disorders. Neurosci Bull 2024; 40:1315-1332. [PMID: 38466557 PMCID: PMC11365908 DOI: 10.1007/s12264-024-01189-z] [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: 08/08/2023] [Accepted: 11/23/2023] [Indexed: 03/13/2024] Open
Abstract
Human pluripotent stem cell (hPSC) models provide unprecedented opportunities to study human neurological disorders by recapitulating human-specific disease mechanisms. In particular, hPSC-based human-animal brain chimeras enable the study of human cell pathophysiology in vivo. In chimeric brains, human neural and immune cells can maintain human-specific features, undergo maturation, and functionally integrate into host brains, allowing scientists to study how human cells impact neural circuits and animal behaviors. The emerging human-animal brain chimeras hold promise for modeling human brain cells and their interactions in health and disease, elucidating the disease mechanism from molecular and cellular to circuit and behavioral levels, and testing the efficacy of cell therapy interventions. Here, we discuss recent advances in the generation and applications of using human-animal chimeric brain models for the study of neurological disorders, including disease modeling and cell therapy.
Collapse
Affiliation(s)
- Yanru Ji
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Jenna Lillie McLean
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Ranjie Xu
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
80
|
Pavlou A, Mulenge F, Gern OL, Busker LM, Greimel E, Waltl I, Kalinke U. Orchestration of antiviral responses within the infected central nervous system. Cell Mol Immunol 2024; 21:943-958. [PMID: 38997413 PMCID: PMC11364666 DOI: 10.1038/s41423-024-01181-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/05/2024] [Indexed: 07/14/2024] Open
Abstract
Many newly emerging and re-emerging viruses have neuroinvasive potential, underscoring viral encephalitis as a global research priority. Upon entry of the virus into the CNS, severe neurological life-threatening conditions may manifest that are associated with high morbidity and mortality. The currently available therapeutic arsenal against viral encephalitis is rather limited, emphasizing the need to better understand the conditions of local antiviral immunity within the infected CNS. In this review, we discuss new insights into the pathophysiology of viral encephalitis, with a focus on myeloid cells and CD8+ T cells, which critically contribute to protection against viral CNS infection. By illuminating the prerequisites of myeloid and T cell activation, discussing new discoveries regarding their transcriptional signatures, and dissecting the mechanisms of their recruitment to sites of viral replication within the CNS, we aim to further delineate the complexity of antiviral responses within the infected CNS. Moreover, we summarize the current knowledge in the field of virus infection and neurodegeneration and discuss the potential links of some neurotropic viruses with certain pathological hallmarks observed in neurodegeneration.
Collapse
Affiliation(s)
- Andreas Pavlou
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Felix Mulenge
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Olivia Luise Gern
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Lena Mareike Busker
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, 30559, Hannover, Germany
| | - Elisabeth Greimel
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Inken Waltl
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany.
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
81
|
Chen X, Huang Y, Huang L, Huang Z, Hao ZZ, Xu L, Xu N, Li Z, Mou Y, Ye M, You R, Zhang X, Liu S, Miao Z. A brain cell atlas integrating single-cell transcriptomes across human brain regions. Nat Med 2024; 30:2679-2691. [PMID: 39095595 PMCID: PMC11405287 DOI: 10.1038/s41591-024-03150-z] [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: 07/31/2023] [Accepted: 06/24/2024] [Indexed: 08/04/2024]
Abstract
While single-cell technologies have greatly advanced our comprehension of human brain cell types and functions, studies including large numbers of donors and multiple brain regions are needed to extend our understanding of brain cell heterogeneity. Integrating atlas-level single-cell data presents a chance to reveal rare cell types and cellular heterogeneity across brain regions. Here we present the Brain Cell Atlas, a comprehensive reference atlas of brain cells, by assembling single-cell data from 70 human and 103 mouse studies of the brain throughout major developmental stages across brain regions, covering over 26.3 million cells or nuclei from both healthy and diseased tissues. Using machine-learning based algorithms, the Brain Cell Atlas provides a consensus cell type annotation, and it showcases the identification of putative neural progenitor cells and a cell subpopulation of PCDH9high microglia in the human brain. We demonstrate the gene regulatory difference of PCDH9high microglia between hippocampus and prefrontal cortex and elucidate the cell-cell communication network. The Brain Cell Atlas presents an atlas-level integrative resource for comparing brain cells in different environments and conditions within the Human Cell Atlas.
Collapse
Affiliation(s)
- Xinyue Chen
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, China
| | - Yin Huang
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, China
| | - Liangfeng Huang
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, China
| | - Ziliang Huang
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, China
| | - Zhao-Zhe Hao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Lahong Xu
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, China
| | - Nana Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Zhi Li
- Department of Neurosurgery/Neuro-oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yonggao Mou
- Department of Neurosurgery/Neuro-oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Mingli Ye
- Tsinghua Fuzhou Institute for Data Technology, Fuzhou, China
| | - Renke You
- Tsinghua Fuzhou Institute for Data Technology, Fuzhou, China
| | - Xuegong Zhang
- MOE Key Lab of Bioinformatics, Bioinformatics Division of BNRIST and Department of Automation, Tsinghua University, Beijing, China
- School of Medicine, Tsinghua University, Beijing, China
- School of Life Sciences, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
| | - Sheng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Guangzhou, China.
| | - Zhichao Miao
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou International Bio Island, Guangzhou, China.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou International Bio Island, Guangzhou, China.
| |
Collapse
|
82
|
Bellotti C, Samudyata S, Thams S, Sellgren CM, Rostami E. Organoids and chimeras: the hopeful fusion transforming traumatic brain injury research. Acta Neuropathol Commun 2024; 12:141. [PMID: 39215375 PMCID: PMC11363608 DOI: 10.1186/s40478-024-01845-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/10/2024] [Indexed: 09/04/2024] Open
Abstract
Research in the field of traumatic brain injury has until now heavily relied on the use of animal models to identify potential therapeutic approaches. However, a long series of failed clinical trials has brought many scientists to question the translational reliability of pre-clinical results obtained in animals. The search for an alternative to conventional models that better replicate human pathology in traumatic brain injury is thus of the utmost importance for the field. Recently, orthotopic xenotransplantation of human brain organoids into living animal models has been achieved. This review summarizes the existing literature on this new method, focusing on its potential applications in preclinical research, both in the context of cell replacement therapy and disease modelling. Given the obvious advantages of this approach to study human pathologies in an in vivo context, we here critically review its current limitations while considering its possible applications in traumatic brain injury research.
Collapse
Affiliation(s)
- Cristina Bellotti
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Samudyata Samudyata
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Sebastian Thams
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Stockholm Health Care Services, Karolinska Institutet, and Stockholm Health Care Services, Stockholm, Sweden
| | - Elham Rostami
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
83
|
Vieira de Sá R, Sudria-Lopez E, Cañizares Luna M, Harschnitz O, van den Heuvel DMA, Kling S, Vonk D, Westeneng HJ, Karst H, Bloemenkamp L, Varderidou-Minasian S, Schlegel DK, Mars M, Broekhoven MH, van Kronenburg NCH, Adolfs Y, Vangoor VR, de Jongh R, Ljubikj T, Peeters L, Seeler S, Mocholi E, Basak O, Gordon D, Giuliani F, Verhoeff T, Korsten G, Calafat Pla T, Venø MT, Kjems J, Talbot K, van Es MA, Veldink JH, van den Berg LH, Zelina P, Pasterkamp RJ. ATAXIN-2 intermediate-length polyglutamine expansions elicit ALS-associated metabolic and immune phenotypes. Nat Commun 2024; 15:7484. [PMID: 39209824 PMCID: PMC11362472 DOI: 10.1038/s41467-024-51676-0] [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: 02/24/2023] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
Intermediate-length repeat expansions in ATAXIN-2 (ATXN2) are the strongest genetic risk factor for amyotrophic lateral sclerosis (ALS). At the molecular level, ATXN2 intermediate expansions enhance TDP-43 toxicity and pathology. However, whether this triggers ALS pathogenesis at the cellular and functional level remains unknown. Here, we combine patient-derived and mouse models to dissect the effects of ATXN2 intermediate expansions in an ALS background. iPSC-derived motor neurons from ATXN2-ALS patients show altered stress granules, neurite damage and abnormal electrophysiological properties compared to healthy control and other familial ALS mutations. In TDP-43Tg-ALS mice, ATXN2-Q33 causes reduced motor function, NMJ alterations, neuron degeneration and altered in vitro stress granule dynamics. Furthermore, gene expression changes related to mitochondrial function and inflammatory response are detected and confirmed at the cellular level in mice and human neuron and organoid models. Together, these results define pathogenic defects underlying ATXN2-ALS and provide a framework for future research into ATXN2-dependent pathogenesis and therapy.
Collapse
Affiliation(s)
- Renata Vieira de Sá
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Emma Sudria-Lopez
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Marta Cañizares Luna
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Oliver Harschnitz
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CX, Utrecht, The Netherlands
- Human Technopole, Viale Rita Levi-Montalcini, 1, 20157, Milan, Italy
| | - Dianne M A van den Heuvel
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Sandra Kling
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Danielle Vonk
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Henk-Jan Westeneng
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CX, Utrecht, The Netherlands
| | - Henk Karst
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Lauri Bloemenkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Suzy Varderidou-Minasian
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Domino K Schlegel
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Mayte Mars
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Mark H Broekhoven
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Nicky C H van Kronenburg
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Youri Adolfs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Vamshidhar R Vangoor
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Rianne de Jongh
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Tijana Ljubikj
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Lianne Peeters
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Sabine Seeler
- Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Enric Mocholi
- Center for Molecuar Medicine, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Onur Basak
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - David Gordon
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, UK
| | - Fabrizio Giuliani
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CX, Utrecht, The Netherlands
| | - Tessa Verhoeff
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Giel Korsten
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Teresa Calafat Pla
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Morten T Venø
- Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
- Omiics ApS, Aarhus, Denmark
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, UK
| | - Michael A van Es
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CX, Utrecht, The Netherlands
| | - Jan H Veldink
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CX, Utrecht, The Netherlands
| | - Leonard H van den Berg
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CX, Utrecht, The Netherlands
| | - Pavol Zelina
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands.
| |
Collapse
|
84
|
Martins-Ferreira R, Calafell-Segura J, Chaves J, Ciudad L, Martins da Silva A, Pinho e Costa P, Leal B, Ballestar E. Purinergic exposure induces epigenomic and transcriptomic-mediated preconditioning resembling epilepsy-associated microglial states. iScience 2024; 27:110546. [PMID: 39184445 PMCID: PMC11342283 DOI: 10.1016/j.isci.2024.110546] [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: 07/10/2023] [Revised: 03/10/2024] [Accepted: 07/16/2024] [Indexed: 08/27/2024] Open
Abstract
Microglia play a crucial role in a range of neuropathologies through exacerbated activation. Microglial inflammatory responses can be influenced by prior exposures to noxious stimuli, like increased levels of extracellular adenosine and ATP. These are characteristic of brain insults like epileptic seizures and could potentially shape subsequent responses through epigenetic regulation. We investigated DNA methylation and expression changes in human microglia-like cells differentiated from monocytes following ATP-mediated preconditioning. We demonstrate that microglia-like cells display homeostatic microglial features, shown by surface markers, transcriptome, and DNA methylome. After exposure to ATP, TLR-mediated activation leads to an exacerbated pro-inflammatory response. These changes are accompanied by methylation and transcriptional reprogramming associated with enhanced immune-related functions. The reprogramming associated with ATP-mediated preconditioning leads to profiles found in microglial subsets linked to epilepsy. Purine-driven microglia immune preconditioning drives epigenetic and transcriptional changes that could contribute to altered functions of microglia during seizure development and progression.
Collapse
Affiliation(s)
- Ricardo Martins-Ferreira
- Epigenetics and Immune Disease Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Spain
- Immunogenetics Laboratory, Molecular Pathology and Immunology Department, Instituto de Ciências Biomédicas Abel Salazar – Universidade do Porto (ICBAS-UPorto), 4050-313 Porto, Portugal
- Autoimmunity and Neuroscience Group. Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
- ITR - Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
| | - Josep Calafell-Segura
- Epigenetics and Immune Disease Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Spain
| | - João Chaves
- Autoimmunity and Neuroscience Group. Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
- ITR - Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
- Neurology Service, Centro Hospitalar Universitário de Santo António (CHUdSA), 4099-001 Porto, Portugal
| | - Laura Ciudad
- Epigenetics and Immune Disease Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Spain
| | - António Martins da Silva
- Autoimmunity and Neuroscience Group. Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
- ITR - Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
- Neurophysiology Service, CHUdSA 4099-001 Porto, Portugal
| | - Paulo Pinho e Costa
- Immunogenetics Laboratory, Molecular Pathology and Immunology Department, Instituto de Ciências Biomédicas Abel Salazar – Universidade do Porto (ICBAS-UPorto), 4050-313 Porto, Portugal
- Autoimmunity and Neuroscience Group. Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
- ITR - Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
- Department of Human Genetics, Instituto Nacional de Saúde Dr. Ricardo Jorge 4000-055 Porto, Portugal
| | - Bárbara Leal
- Immunogenetics Laboratory, Molecular Pathology and Immunology Department, Instituto de Ciências Biomédicas Abel Salazar – Universidade do Porto (ICBAS-UPorto), 4050-313 Porto, Portugal
- Autoimmunity and Neuroscience Group. Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
- ITR - Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
| | - Esteban Ballestar
- Epigenetics and Immune Disease Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Spain
- Epigenetics in Inflammatory and Metabolic Diseases Laboratory, Health Science Center (HSC), East China Normal University (ECNU), Shanghai 200241, China
| |
Collapse
|
85
|
Wenzel TJ, Desjarlais JD, Mousseau DD. Human brain organoids containing microglia that have arisen innately adapt to a β-amyloid challenge better than those in which microglia are integrated by co-culture. Stem Cell Res Ther 2024; 15:258. [PMID: 39135132 PMCID: PMC11320858 DOI: 10.1186/s13287-024-03876-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 08/01/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Alzheimer disease (AD) is a heterogenous and multifactorial disease, and its pathology is partly driven by microglia and their activated phenotype. Brain organoids (BOs) are gaining prominence as a relevant model of the human brain for the study of AD; however, BOs are commonly devoid of microglia. To overcome this limitation, current protocols incorporate microglia through either (1) co-culture (BO co-culture), or (2) molecular manipulation at critical windows of BO development to have microglia arise innately (BO innate cultures). It is currently unclear whether the microglia incorporated into BOs by either of these two protocols differ in function. METHODS At in vitro day 90, BO innate cultures and BO-co-cultures were challenged with the AD-related β-amyloid peptide (Aβ) for up to 72 h. After Aβ challenge, BOs were collected for immunoblotting. Immunoblots compared immunodensity and protein banding of Aβ and ionized calcium-binding adapter molecule 1 (IBA1, a marker of microglial activation) in BOs. The translational potential of these observations was supported using 56 human cortical samples from neurocognitively normal donors and patients with early-onset AD and late-onset AD. Statistical analyses were conducted using the Kruskal-Wallis test, a two-way ANOVA, or a simple linear regression, and where applicable, followed by Dunn's or Sidak's test. RESULTS We show that BO co-cultures promote Aβ oligomerization as early as 24 h and this coincides with a significant increase in IBA1 levels. In contrast, the Aβs do not oligomerize in BO innate cultures and the IBA1 response was modest and only emerged after 48 h. In human cortical samples, we found IBA1 levels correlated with age at onset, age at death, and the putative diagnostic Aβ(1-42)/Aβ(1-40) ratio (particularly in their oligomeric forms) in a sex-dependent manner. CONCLUSIONS Our unique observations suggest that BOs with innate microglia model the response of a healthy brain to Aβ, and by extension the initial stages of Aβ challenge. It would be impossible to model these early stages of pathogenesis in BOs where microglia are already compromised, such as those with microglia incorporated by co-culture.
Collapse
Affiliation(s)
- Tyler J Wenzel
- Cell Signalling Laboratory, Department of Psychiatry, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada.
| | - Joseph D Desjarlais
- Cell Signalling Laboratory, Department of Psychiatry, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Darrell D Mousseau
- Cell Signalling Laboratory, Department of Psychiatry, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| |
Collapse
|
86
|
Vicario R, Fragkogianni S, Weber L, Lazarov T, Hu Y, Hayashi SY, Craddock BP, Socci ND, Alberdi A, Baako A, Ay O, Ogishi M, Lopez-Rodrigo E, Kappagantula R, Viale A, Iacobuzio-Donahue CA, Zhou T, Ransohoff RM, Chesworth R, Netherlands Brain Bank, Abdel-Wahab O, Boisson B, Elemento O, Casanova JL, Miller WT, Geissmann F. A microglia clonal inflammatory disorder in Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.25.577216. [PMID: 38328106 PMCID: PMC10849735 DOI: 10.1101/2024.01.25.577216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Somatic genetic heterogeneity resulting from post-zygotic DNA mutations is widespread in human tissues and can cause diseases, however few studies have investigated its role in neurodegenerative processes such as Alzheimer's Disease (AD). Here we report the selective enrichment of microglia clones carrying pathogenic variants, that are not present in neuronal, glia/stromal cells, or blood, from patients with AD in comparison to age-matched controls. Notably, microglia-specific AD-associated variants preferentially target the MAPK pathway, including recurrent CBL ring-domain mutations. These variants activate ERK and drive a microglia transcriptional program characterized by a strong neuro-inflammatory response, both in vitro and in patients. Although the natural history of AD-associated microglial clones is difficult to establish in human, microglial expression of a MAPK pathway activating variant was previously shown to cause neurodegeneration in mice, suggesting that AD-associated neuroinflammatory microglial clones may contribute to the neurodegenerative process in patients.
Collapse
Affiliation(s)
- Rocio Vicario
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Stamatina Fragkogianni
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Leslie Weber
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Tomi Lazarov
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Yang Hu
- Department of Physiology and Biophysics, Institute for Computational Biomedicine,Weill Cornell New York, NY 10021, USA
| | - Samantha Y. Hayashi
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY, 11794-8661
| | - Barbara P. Craddock
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY, 11794-8661
| | - Nicholas D. Socci
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Araitz Alberdi
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Ann Baako
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Oyku Ay
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, 10065 NY, USA
| | - Estibaliz Lopez-Rodrigo
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Rajya Kappagantula
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Agnes Viale
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Christine A. Iacobuzio-Donahue
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Ting Zhou
- SKI Stem Cell Research Core, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | | | | | | | - Omar Abdel-Wahab
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Bertrand Boisson
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, 10065 NY, USA
| | - Olivier Elemento
- Department of Physiology and Biophysics, Institute for Computational Biomedicine,Weill Cornell New York, NY 10021, USA
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, 10065 NY, USA
| | - W. Todd Miller
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY, 11794-8661
| | - Frederic Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| |
Collapse
|
87
|
Fernandes DC, Eto SF, Baldassi AC, Balbuena TS, Charlie-Silva I, de Andrade Belo MA, Pizauro JM. Meningitis caused by Aeromonas hydrophila in Oreochromis niloticus: Proteomics and druggability of virulence factors. FISH & SHELLFISH IMMUNOLOGY 2024; 151:109687. [PMID: 38866348 DOI: 10.1016/j.fsi.2024.109687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/30/2024] [Accepted: 06/08/2024] [Indexed: 06/14/2024]
Abstract
Meningitis caused by Gram-negative bacteria is a serious public health problem, causing morbidity and mortality in both children and adults. Here, we propose a novel experimental model using Nile tilapia (Oreochromis niloticus) to study neuroinflammation. The fish were infected with Aeromonas hydrophila, and the course of infection was monitored in the peripheral blood. Septicemia was obvious in the blood, while in the brain tissue, infection of the meninges was present. The histopathological examination showed suppurative meningitis, and the cellular immune response in the brain tissue during infection was mediated by microglia. These cells were morphologically characterized and phenotyped by MHC class II markers and CD68. The increased production of TNF-α, IL-1β and iNOS supported the infiltration of these cells during the neuroinflammatory process. In the proteomic analysis of A. hydrophila isolated from brain tissue, we found chemotactic and transport proteins, proteolytic enzymes and enzymes associated with the dismutation of nitric oxide (NO), as well as motor proteins and those responsible for cell division. After characterizing the most abundant proteins during the course of infection, we investigated the druggability index of these proteins and identified promising peptide sequences as molecular targets that are similar among bacteria. Thus, these findings deepened the understanding of the pathophysiology of meningitis caused by A. hydrophila. Moreover, through the proteomics analysis, important mechanisms and pathways used by the pathogen to subvert the host response were revealed, providing insights for the development of novel antibiotics and vaccines.
Collapse
Affiliation(s)
- Dayanne Carla Fernandes
- Institute of Chemistry, São Paulo State University (Unesp), Araraquara, Sao Paulo, SP, Brazil.
| | - Silas Fernandes Eto
- Laboratory Center of Excellence in New Target Discovery (CENTD) Special Laboratory, Butantan Institute, São Paulo, SP, Brazil
| | - Amanda Cristina Baldassi
- Department of Technology, School of Agrarian and Veterinary Sciences, São Paulo State University (Unesp), Jaboticabal, Sao Paulo, SP, Brazil
| | - Thiago Santana Balbuena
- Department of Technology, School of Agrarian and Veterinary Sciences, São Paulo State University (Unesp), Jaboticabal, Sao Paulo, SP, Brazil
| | - Ives Charlie-Silva
- Institute of Chemistry, São Paulo State University (Unesp), Araraquara, Sao Paulo, SP, Brazil
| | | | - João Martins Pizauro
- Department of Technology, School of Agrarian and Veterinary Sciences, São Paulo State University (Unesp), Jaboticabal, Sao Paulo, SP, Brazil
| |
Collapse
|
88
|
Sierra A, Miron VE, Paolicelli RC, Ransohoff RM. Microglia in Health and Diseases: Integrative Hubs of the Central Nervous System (CNS). Cold Spring Harb Perspect Biol 2024; 16:a041366. [PMID: 38438189 PMCID: PMC11293550 DOI: 10.1101/cshperspect.a041366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Microglia are usually referred to as "the innate immune cells of the brain," "the resident macrophages of the central nervous system" (CNS), or "CNS parenchymal macrophages." These labels allude to their inherent immune function, related to their macrophage lineage. However, beyond their classic innate immune responses, microglia also play physiological roles crucial for proper brain development and maintenance of adult brain homeostasis. Microglia sense both external and local stimuli through a variety of surface receptors. Thus, they might serve as integrative hubs at the interface between the external environment and the CNS, able to decode, filter, and buffer cues from outside, with the aim of preserving and maintaining brain homeostasis. In this perspective, we will cast a critical look at how these multiple microglial functions are acquired and coordinated, and we will speculate on their impact on human brain physiology and pathology.
Collapse
Affiliation(s)
- Amanda Sierra
- Achucarro Basque Center for Neuroscience, Glial Cell Biology Laboratory, Science Park of UPV/EHU, E-48940 Leioa, Bizkaia, Spain
- Department of Biochemistry and Molecular Biology, University of the Basque Country EHU/UPV, 48940 Leioa, Spain
- Ikerbasque Foundation, Bilbao 48009, Spain
| | - Veronique E Miron
- BARLO Multiple Sclerosis Centre, Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto M5B 1T8, Canada
- Department of Immunology, University of Toronto, Toronto M5S 1A8, Canada
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4TJ, United Kingdom
| | - Rosa C Paolicelli
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, CH-1005 Lausanne, Switzerland
| | | |
Collapse
|
89
|
Pramotton FM, Spitz S, Kamm RD. Challenges and Future Perspectives in Modeling Neurodegenerative Diseases Using Organ-on-a-Chip Technology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403892. [PMID: 38922799 PMCID: PMC11348103 DOI: 10.1002/advs.202403892] [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: 04/12/2024] [Revised: 06/01/2024] [Indexed: 06/28/2024]
Abstract
Neurodegenerative diseases (NDDs) affect more than 50 million people worldwide, posing a significant global health challenge as well as a high socioeconomic burden. With aging constituting one of the main risk factors for some NDDs such as Alzheimer's disease (AD) and Parkinson's disease (PD), this societal toll is expected to rise considering the predicted increase in the aging population as well as the limited progress in the development of effective therapeutics. To address the high failure rates in clinical trials, legislative changes permitting the use of alternatives to traditional pre-clinical in vivo models are implemented. In this regard, microphysiological systems (MPS) such as organ-on-a-chip (OoC) platforms constitute a promising tool, due to their ability to mimic complex and human-specific tissue niches in vitro. This review summarizes the current progress in modeling NDDs using OoC technology and discusses five critical aspects still insufficiently addressed in OoC models to date. Taking these aspects into consideration in the future MPS will advance the modeling of NDDs in vitro and increase their translational value in the clinical setting.
Collapse
Affiliation(s)
- Francesca Michela Pramotton
- Department of Mechanical Engineering and Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Sarah Spitz
- Department of Mechanical Engineering and Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Roger D. Kamm
- Department of Mechanical Engineering and Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| |
Collapse
|
90
|
Ramamurthy E, Agarwal S, Toong N, Sestili H, Kaplow IM, Chen Z, Phan B, Pfenning AR. Regression convolutional neural network models implicate peripheral immune regulatory variants in the predisposition to Alzheimer's disease. PLoS Comput Biol 2024; 20:e1012356. [PMID: 39186798 PMCID: PMC11389932 DOI: 10.1371/journal.pcbi.1012356] [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: 06/24/2023] [Revised: 09/11/2024] [Accepted: 07/23/2024] [Indexed: 08/28/2024] Open
Abstract
Alzheimer's disease (AD) involves aggregation of amyloid β and tau, neuron loss, cognitive decline, and neuroinflammatory responses. Both resident microglia and peripheral immune cells have been associated with the immune component of AD. However, the relative contribution of resident and peripheral immune cell types to AD predisposition has not been thoroughly explored due to their similarity in gene expression and function. To study the effects of AD-associated variants on cis-regulatory elements, we train convolutional neural network (CNN) regression models that link genome sequence to cell type-specific levels of open chromatin, a proxy for regulatory element activity. We then use in silico mutagenesis of regulatory sequences to predict the relative impact of candidate variants across these cell types. We develop and apply criteria for evaluating our models and refine our models using massively parallel reporter assay (MPRA) data. Our models identify multiple AD-associated variants with a greater predicted impact in peripheral cells relative to microglia or neurons. Our results support their use as models to study the effects of AD-associated variants and even suggest that peripheral immune cells themselves may mediate a component of AD predisposition. We make our library of CNN models and predictions available as a resource for the community to study immune and neurological disorders.
Collapse
Affiliation(s)
- Easwaran Ramamurthy
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Snigdha Agarwal
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Noelle Toong
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Heather Sestili
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Irene M. Kaplow
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Ziheng Chen
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - BaDoi Phan
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Andreas R. Pfenning
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
91
|
Carrier M, Robert MÈ, St-Pierre MK, Ibáñez FG, Gonçalves de Andrade E, Laroche A, Picard K, Vecchiarelli HA, Savage JC, Boilard É, Desjardins M, Tremblay MÈ. Bone marrow-derived myeloid cells transiently colonize the brain during postnatal development and interact with glutamatergic synapses. iScience 2024; 27:110037. [PMID: 39021809 PMCID: PMC11253522 DOI: 10.1016/j.isci.2024.110037] [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: 10/09/2023] [Revised: 03/01/2024] [Accepted: 05/16/2024] [Indexed: 07/20/2024] Open
Abstract
Although the roles of embryonic yolk sac-derived, resident microglia in neurodevelopment were extensively studied, the possible involvement of bone marrow-derived cells remains elusive. In this work, we used a fate-mapping strategy to selectively label bone marrow-derived cells and their progeny in the brain (FLT3+IBA1+). FLT3+IBA1+ cells were confirmed to be transiently present in the healthy brain during early postnatal development. FLT3+IBA1+ cells have a distinct morphology index at postnatal day(P)0, P7, and P14 compared with neighboring microglia. FLT3+IBA1+ cells also express the microglial markers P2RY12 and TMEM119 and interact with VGLUT1 synapses at P14. Scanning electron microscopy indeed showed that FLT3+ cells contact and engulf pre-synaptic elements. Our findings suggest FLT3+IBA1+ cells might assist microglia in their physiological functions in the developing brain including synaptic pruning which is performed using their purinergic sensors. Our findings stimulate further investigation on the involvement of peripheral macrophages during homeostatic and pathological development.
Collapse
Affiliation(s)
- Micaël Carrier
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
- Département de psychiatrie et de neurosciences, Faculté de médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 3E6, Canada
| | - Marie-Ève Robert
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
| | - Marie-Kim St-Pierre
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 3E6, Canada
- Département de médecine moléculaire, Faculté de médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Fernando González Ibáñez
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 3E6, Canada
- Département de médecine moléculaire, Faculté de médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | | | - Audrée Laroche
- Département de microbiologie et immunologie, Faculté de médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Katherine Picard
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 3E6, Canada
| | | | - Julie C. Savage
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
| | - Éric Boilard
- Département de microbiologie et immunologie, Faculté de médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Michèle Desjardins
- Department of Physics, Physical Engineering and Optics, Université Laval, Québec City, QC G1V 0A6, Canada
- Oncology Division, Centre de recherche du CHU de Québec, Université Laval, Québec City, QC G1V 4G2, Canada
| | - Marie-Ève Tremblay
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 3E6, Canada
- Département de médecine moléculaire, Faculté de médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC V6T 1Z4 Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC H3A 0G4 Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, BC V8W 2Y2, Canada
| |
Collapse
|
92
|
Nevelchuk S, Brawek B, Schwarz N, Valiente-Gabioud A, Wuttke TV, Kovalchuk Y, Koch H, Höllig A, Steiner F, Figarella K, Griesbeck O, Garaschuk O. Morphotype-specific calcium signaling in human microglia. J Neuroinflammation 2024; 21:175. [PMID: 39020359 PMCID: PMC11256502 DOI: 10.1186/s12974-024-03169-6] [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: 05/25/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Key functions of Ca2+ signaling in rodent microglia include monitoring the brain state as well as the surrounding neuronal activity and sensing the danger or damage in their vicinity. Microglial Ca2+ dyshomeostasis is a disease hallmark in many mouse models of neurological disorders but the Ca2+ signal properties of human microglia remain unknown. METHODS We developed a novel genetically-encoded ratiometric Ca2+ indicator, targeting microglial cells in the freshly resected human tissue, organotypically cultured tissue slices and analyzed in situ ongoing Ca2+ signaling of decades-old microglia dwelling in their native microenvironment. RESULTS The data revealed marked compartmentalization of Ca2+ signals, with signal properties differing across the compartments and resident morphotypes. The basal Ca2+ levels were low in ramified and high in ameboid microglia. The fraction of cells with ongoing Ca2+ signaling, the fraction and the amplitude of process Ca2+ signals and the duration of somatic Ca2+ signals decreased when moving from ramified via hypertrophic to ameboid microglia. In contrast, the size of active compartments, the fraction and amplitude of somatic Ca2+ signals and the duration of process Ca2+ signals increased along this pathway.
Collapse
Affiliation(s)
- Sofia Nevelchuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
| | - Bianca Brawek
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
| | - Niklas Schwarz
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Ariel Valiente-Gabioud
- Tools for Bio-Imaging, Max-Planck-Institute for Biological Intelligence, Martinsried, Germany
| | - Thomas V Wuttke
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Department of Neurosurgery, University of Tübingen, Tübingen, Germany
| | - Yury Kovalchuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
| | - Henner Koch
- Department of Epileptology, Neurology, RWTH Aachen University Hospital, Aachen, Germany
| | - Anke Höllig
- Department of Neurosurgery, RWTH Aachen University, Aachen, Germany
| | - Frederik Steiner
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
| | - Katherine Figarella
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Oliver Griesbeck
- Tools for Bio-Imaging, Max-Planck-Institute for Biological Intelligence, Martinsried, Germany
| | - Olga Garaschuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany.
| |
Collapse
|
93
|
Diaz-Salazar C, Krzisch M, Yoo J, Nano PR, Bhaduri A, Jaenisch R, Polleux F. Human-specific paralogs of SRGAP2 induce neotenic features of microglia structural and functional maturation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601266. [PMID: 38979266 PMCID: PMC11230448 DOI: 10.1101/2024.06.28.601266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Microglia play key roles in shaping synaptic connectivity during neural circuits development. Whether microglia display human-specific features of structural and functional maturation is currently unknown. We show that the ancestral gene SRGAP2A and its human-specific (HS) paralogs SRGAP2B/C are not only expressed in cortical neurons but are the only HS gene duplications expressed in human microglia. Here, using combination of xenotransplantation of human induced pluripotent stem cell (hiPSC)-derived microglia and mouse genetic models, we demonstrate that (1) HS SRGAP2B/C are necessary and sufficient to induce neotenic features of microglia structural and functional maturation in a cell-autonomous manner, and (2) induction of SRGAP2-dependent neotenic features of microglia maturation non-cell autonomously impacts synaptic development in cortical pyramidal neurons. Our results reveal that, during human brain evolution, human-specific genes SRGAP2B/C coordinated the emergence of neotenic features of synaptic development by acting as genetic modifiers of both neurons and microglia.
Collapse
Affiliation(s)
- Carlos Diaz-Salazar
- Department of Neuroscience, Columbia University, New York, NY, 10027, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University; New York, NY, 10027, USA
| | - Marine Krzisch
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Juyoun Yoo
- Department of Neuroscience, Columbia University, New York, NY, 10027, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University; New York, NY, 10027, USA
| | - Patricia R. Nano
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Aparna Bhaduri
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Franck Polleux
- Department of Neuroscience, Columbia University, New York, NY, 10027, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University; New York, NY, 10027, USA
| |
Collapse
|
94
|
Berki P, Cserép C, Környei Z, Pósfai B, Szabadits E, Domonkos A, Kellermayer A, Nyerges M, Wei X, Mody I, Kunihiko A, Beck H, Kaikai H, Ya W, Lénárt N, Wu Z, Jing M, Li Y, Gulyás AI, Dénes Á. Microglia contribute to neuronal synchrony despite endogenous ATP-related phenotypic transformation in acute mouse brain slices. Nat Commun 2024; 15:5402. [PMID: 38926390 PMCID: PMC11208608 DOI: 10.1038/s41467-024-49773-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Acute brain slices represent a workhorse model for studying the central nervous system (CNS) from nanoscale events to complex circuits. While slice preparation inherently involves tissue damage, it is unclear how microglia, the main immune cells and damage sensors of the CNS react to this injury and shape neuronal activity ex vivo. To this end, we investigated microglial phenotypes and contribution to network organization and functioning in acute brain slices. We reveal time-dependent microglial phenotype changes influenced by complex extracellular ATP dynamics through P2Y12R and CX3CR1 signalling, which is sustained for hours in ex vivo mouse brain slices. Downregulation of P2Y12R and changes of microglia-neuron interactions occur in line with alterations in the number of excitatory and inhibitory synapses over time. Importantly, functional microglia modulate synapse sprouting, while microglial dysfunction results in markedly impaired ripple activity both ex vivo and in vivo. Collectively, our data suggest that microglia are modulators of complex neuronal networks with important roles to maintain neuronal network integrity and activity. We suggest that slice preparation can be used to model time-dependent changes of microglia-neuron interactions to reveal how microglia shape neuronal circuits in physiological and pathological conditions.
Collapse
Affiliation(s)
- Péter Berki
- János Szentágothai Doctoral School of Neuroscience, Semmelweis University, Budapest, H-1083, Hungary
- Laboratory of Cerebral Cortex Research, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
- Laboratory of Neuronal Network and Behaviour, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Csaba Cserép
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Zsuzsanna Környei
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Balázs Pósfai
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Eszter Szabadits
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Andor Domonkos
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
- Laboratory of Thalamus Research, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Anna Kellermayer
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Miklós Nyerges
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Xiaofei Wei
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Istvan Mody
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Araki Kunihiko
- Institute of Experimental Epileptology and Cognition Research, Medical University of Bonn, Bonn, 53127, Germany
- University Hospital Bonn, Bonn, Germany
| | - Heinz Beck
- Institute of Experimental Epileptology and Cognition Research, Medical University of Bonn, Bonn, 53127, Germany
- University Hospital Bonn, Bonn, Germany
| | - He Kaikai
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, School of Life Sciences, Peking University, 100871, Beijing, China
| | - Wang Ya
- Chinese Institute for Brain Research, 102206, Beijing, China
| | - Nikolett Lénárt
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Zhaofa Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Miao Jing
- Chinese Institute for Brain Research, 102206, Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, School of Life Sciences, Peking University, 100871, Beijing, China
| | - Attila I Gulyás
- Laboratory of Cerebral Cortex Research, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Ádám Dénes
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary.
| |
Collapse
|
95
|
Ghosh Roy S, Karim AF, Brumeanu TD, Casares SA. Reconstitution of human microglia and resident T cells in the brain of humanized DRAGA mice. Front Cell Infect Microbiol 2024; 14:1367566. [PMID: 38983114 PMCID: PMC11231403 DOI: 10.3389/fcimb.2024.1367566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 06/10/2024] [Indexed: 07/11/2024] Open
Abstract
Humanized mouse models are valuable tools for investigating the human immune system in response to infection and injury. We have previously described the human immune system (HIS)-DRAGA mice (HLA-A2.HLA-DR4.Rag1KO.IL-2RgKO.NOD) generated by infusion of Human Leukocyte Antigen (HLA)-matched, human hematopoietic stem cells from umbilical cord blood. By reconstituting human cells, the HIS-DRAGA mouse model has been utilized as a "surrogate in vivo human model" for infectious diseases such as Human Immunodeficiency Virus (HIV), Influenza, Coronavirus Disease 2019 (COVID-19), scrub typhus, and malaria. This humanized mouse model bypasses ethical concerns about the use of fetal tissues for the humanization of laboratory animals. Here in, we demonstrate the presence of human microglia and T cells in the brain of HIS-DRAGA mice. Microglia are brain-resident macrophages that play pivotal roles against pathogens and cerebral damage, whereas the brain-resident T cells provide surveillance and defense against infections. Our findings suggest that the HIS-DRAGA mouse model offers unique advantages for studying the functions of human microglia and T cells in the brain during infections, degenerative disorders, tumors, and trauma, as well as for testing therapeutics in these pathological conditions.
Collapse
Affiliation(s)
- Sounak Ghosh Roy
- Agile Vaccines & Therapeutics, Defense Infectious Diseases Directorate, Naval Medical Research Command, Silver Spring, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Ahmad F. Karim
- Agile Vaccines & Therapeutics, Defense Infectious Diseases Directorate, Naval Medical Research Command, Silver Spring, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Teodor-D. Brumeanu
- Department of Medicine, Division of Immunology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Sofia A. Casares
- Agile Vaccines & Therapeutics, Defense Infectious Diseases Directorate, Naval Medical Research Command, Silver Spring, MD, United States
| |
Collapse
|
96
|
Batorsky R, Ceasrine AM, Shook LL, Kislal S, Bordt EA, Devlin BA, Perlis RH, Slonim DK, Bilbo SD, Edlow AG. Hofbauer cells and fetal brain microglia share transcriptional profiles and responses to maternal diet-induced obesity. Cell Rep 2024; 43:114326. [PMID: 38848212 PMCID: PMC11808824 DOI: 10.1016/j.celrep.2024.114326] [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: 12/13/2023] [Revised: 04/25/2024] [Accepted: 05/22/2024] [Indexed: 06/09/2024] Open
Abstract
Maternal immune activation is associated with adverse offspring neurodevelopmental outcomes, many mediated by in utero microglial programming. As microglia remain inaccessible throughout development, identification of noninvasive biomarkers reflecting fetal brain microglial programming could permit screening and intervention. We used lineage tracing to demonstrate the shared ontogeny between fetal brain macrophages (microglia) and fetal placental macrophages (Hofbauer cells) in a mouse model of maternal diet-induced obesity, and single-cell RNA-seq to demonstrate shared transcriptional programs. Comparison with human datasets demonstrated conservation of placental resident macrophage signatures between mice and humans. Single-cell RNA-seq identified common alterations in fetal microglial and Hofbauer cell gene expression induced by maternal obesity, as well as sex differences in these alterations. We propose that Hofbauer cells, which are easily accessible at birth, provide insights into fetal brain microglial programs and may facilitate the early identification of offspring vulnerable to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Rebecca Batorsky
- Data Intensive Studies Center, Tufts University, Medford, MA, USA
| | - Alexis M Ceasrine
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Lydia L Shook
- Division of Maternal-Fetal Medicine, Department of Ob/Gyn, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, MA, USA
| | - Sezen Kislal
- Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, MA, USA
| | - Evan A Bordt
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Benjamin A Devlin
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Roy H Perlis
- Department of Psychiatry and Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Donna K Slonim
- Department of Computer Science, Tufts University, Medford, MA, USA
| | - Staci D Bilbo
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA; Department of Neurobiology, Duke University, Durham, NC, USA; Lurie Center for Autism, Massachusetts General Hospital, Boston, MA, USA
| | - Andrea G Edlow
- Division of Maternal-Fetal Medicine, Department of Ob/Gyn, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
97
|
Matt SM, Nolan R, Manikandan S, Agarwal Y, Channer B, Oteju O, Daniali M, Canagarajah JA, LuPone T, Mompho K, Runner K, Nickoloff-Bybel E, Li B, Niu M, Schlachetzki JCM, Fox HS, Gaskill PJ. Dopamine-driven Increase in IL-1β in Myeloid Cells is Mediated by Differential Dopamine Receptor Expression and Exacerbated by HIV. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.09.598137. [PMID: 38915663 PMCID: PMC11195146 DOI: 10.1101/2024.06.09.598137] [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
The catecholamine neurotransmitter dopamine is classically known for regulation of central nervous system (CNS) functions such as reward, movement, and cognition. Increasing evidence also indicates that dopamine regulates critical functions in peripheral organs and is an important immunoregulatory factor. We have previously shown that dopamine increases NF-κB activity, inflammasome activation, and the production of inflammatory cytokines such as IL-1β in human macrophages. As myeloid lineage cells are central to the initiation and resolution of acute inflammatory responses, dopamine-mediated dysregulation of these functions could both impair the innate immune response and exacerbate chronic inflammation. However, the exact pathways by which dopamine drives myeloid inflammation are not well defined, and studies in both rodent and human systems indicate that dopamine can impact the production of inflammatory mediators through both D1-like dopamine receptors (DRD1, DRD5) and D2-like dopamine receptors (DRD2, DRD3, and DRD4). Therefore, we hypothesized that dopamine-mediated production of IL-1β in myeloid cells is regulated by the ratio of different dopamine receptors that are activated. Our data in primary human monocyte-derived macrophages (hMDM) indicate that DRD1 expression is necessary for dopamine-mediated increases in IL-1β, and that changes in the expression of DRD2 and other dopamine receptors can alter the magnitude of the dopamine-mediated increase in IL-1β. Mature hMDM have a high D1-like to D2-like receptor ratio, which is different relative to monocytes and peripheral blood mononuclear cells (PBMCs). We further confirm in human microglia cell lines that a high ratio of D1-like to D2-like receptors promotes dopamine-induced increases in IL-1β gene and protein expression using pharmacological inhibition or overexpression of dopamine receptors. RNA-sequencing of dopamine-treated microglia shows that genes encoding functions in IL-1β signaling pathways, microglia activation, and neurotransmission increased with dopamine treatment. Finally, using HIV as an example of a chronic inflammatory disease that is substantively worsened by comorbid substance use disorders (SUDs) that impact dopaminergic signaling, we show increased effects of dopamine on inflammasome activation and IL-1β in the presence of HIV in both human macrophages and microglia. These data suggest that use of addictive substances and dopamine-modulating therapeutics could dysregulate the innate inflammatory response and exacerbate chronic neuroimmunological conditions like HIV. Thus, a detailed understanding of dopamine-mediated changes in inflammation, in particular pathways regulating IL-1β, will be critical to effectively tailor medication regimens.
Collapse
|
98
|
Holt M, Lin J, Cicka M, Wong A, Epelman S, Lavine KJ. Dissecting and Visualizing the Functional Diversity of Cardiac Macrophages. Circ Res 2024; 134:1791-1807. [PMID: 38843293 DOI: 10.1161/circresaha.124.323817] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024]
Abstract
Cardiac macrophages represent a functionally diverse population of cells involved in cardiac homeostasis, repair, and remodeling. With recent advancements in single-cell technologies, it is possible to elucidate specific macrophage subsets based on transcriptional signatures and cell surface protein expression to gain a deep understanding of macrophage diversity in the heart. The use of fate-mapping technologies and parabiosis studies have provided insight into the ontogeny and dynamics of macrophages identifying subsets derived from embryonic and adult definitive hematopoietic progenitors that include tissue-resident and bone marrow monocyte-derived macrophages, respectively. Within the heart, these subsets have distinct tissue niches and functional roles in the setting of homeostasis and disease, with cardiac resident macrophages representing a protective cell population while bone marrow monocyte-derived cardiac macrophages have a context-dependent effect, triggering both proinflammatory tissue injury, but also promoting reparative functions. With the increased understanding of the clinical relevance of cardiac macrophage subsets, there has been an increasing need to detect and measure cardiac macrophage compositions in living animals and patients. New molecular tracers compatible with positron emission tomography/computerized tomography and positron emission tomography/ magnetic resonance imaging have enabled investigators to noninvasively and serially visualize cardiac macrophage subsets within the heart to define associations with disease and measure treatment responses. Today, advancements within this thriving field are poised to fuel an era of clinical translation.
Collapse
Affiliation(s)
- Megan Holt
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine (M.H., M.C., K.J.L.)
| | - Julia Lin
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (J.L., A.W., S.E.)
- Department of Immunology, University of Toronto, ON, Canada (J.L., A.W., S.E.)
| | - Markus Cicka
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine (M.H., M.C., K.J.L.)
| | - Anthony Wong
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (J.L., A.W., S.E.)
- Department of Immunology, University of Toronto, ON, Canada (J.L., A.W., S.E.)
| | - Slava Epelman
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (J.L., A.W., S.E.)
- Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, ON, Canada (S.E.)
- Department of Immunology, University of Toronto, ON, Canada (J.L., A.W., S.E.)
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada (S.E.)
| | - Kory J Lavine
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine (M.H., M.C., K.J.L.)
| |
Collapse
|
99
|
Tharp ME, Han CZ, Balak CD, Fitzpatrick C, O'Connor C, Preissl S, Buchanan J, Nott A, Escoubet L, Mavrommatis K, Gupta M, Schwartz MS, Sang UH, Jones PS, Levy ML, Gonda DD, Ben-Haim S, Ciacci J, Barba D, Khalessi A, Coufal NG, Chen CC, Glass CK, Page DC. The inactive X chromosome drives sex differences in microglial inflammatory activity in human glioblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597433. [PMID: 38895459 PMCID: PMC11185629 DOI: 10.1101/2024.06.06.597433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Biological sex is an important risk factor in cancer, but the underlying cell types and mechanisms remain obscure. Since tumor development is regulated by the immune system, we hypothesize that sex-biased immune interactions underpin sex differences in cancer. The male-biased glioblastoma multiforme (GBM) is an aggressive and treatment-refractory tumor in urgent need of more innovative approaches, such as considering sex differences, to improve outcomes. GBM arises in the specialized brain immune environment dominated by microglia, so we explored sex differences in this immune cell type. We isolated adult human TAM-MGs (tumor-associated macrophages enriched for microglia) and control microglia and found sex-biased inflammatory signatures in GBM and lower-grade tumors associated with pro-tumorigenic activity in males and anti-tumorigenic activity in females. We demonstrated that genes expressed or modulated by the inactive X chromosome facilitate this bias. Together, our results implicate TAM-MGs, specifically their sex chromosomes, as drivers of male bias in GBM.
Collapse
Affiliation(s)
- Marla E Tharp
- Whitehead Institute, Cambridge, MA 02142, USA
- These authors contributed equally
| | - Claudia Z Han
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- These authors contributed equally
| | - Chris D Balak
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Conor Fitzpatrick
- Flow Cytometry Core Facility, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Carolyn O'Connor
- Flow Cytometry Core Facility, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Sebastian Preissl
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- Center for Epigenomics, University of California, San Diego, La Jolla, CA 92093, USA
- Present address: Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Justin Buchanan
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- Center for Epigenomics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Alexi Nott
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Brain Sciences, Imperial College London, London, United Kingdom
- UK Dementia Research Institute, Imperial College London, London, United Kingdom
| | | | | | - Mihir Gupta
- Department of Neurosurgery, University of California, San Diego, La Jolla, CA 92037, USA
- Present address: Department of Neurosurgery, Yale University, New Haven, CT 06520, USA
| | - Marc S Schwartz
- Department of Neurosurgery, University of California, San Diego, La Jolla, CA 92037, USA
| | - U Hoi Sang
- Department of Neurosurgery, University of California, San Diego, La Jolla, CA 92037, USA
| | - Pamela S Jones
- Department of Neurosurgery, University of California, San Diego, La Jolla, CA 92037, USA
- Present address: Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Michael L Levy
- Department of Neurosurgery, University of California, San Diego-Rady Children's Hospital, San Diego, CA 92123, USA
| | - David D Gonda
- Department of Neurosurgery, University of California, San Diego-Rady Children's Hospital, San Diego, CA 92123, USA
| | - Sharona Ben-Haim
- Department of Neurosurgery, University of California, San Diego, La Jolla, CA 92037, USA
| | - Joseph Ciacci
- Department of Neurosurgery, University of California, San Diego, La Jolla, CA 92037, USA
| | - David Barba
- Department of Neurosurgery, University of California, San Diego, La Jolla, CA 92037, USA
| | - Alexander Khalessi
- Department of Neurosurgery, University of California, San Diego, La Jolla, CA 92037, USA
| | - Nicole G Coufal
- Department of Pediatrics University of California, San Diego, La Jolla, CA 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Clark C Chen
- Department of Neurosurgery, University of California, San Diego, La Jolla, CA 92037, USA
- Present address: Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - David C Page
- Whitehead Institute, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA 02142, USA
| |
Collapse
|
100
|
Guillet S, Lazarov T, Jordan N, Boisson B, Tello M, Craddock B, Zhou T, Nishi C, Bareja R, Yang H, Rieux-Laucat F, Lorenzo RIF, Dyall SD, Isenberg D, D’Cruz D, Lachmann N, Elemento O, Viale A, Socci ND, Abel L, Nagata S, Huse M, Miller WT, Casanova JL, Geissmann F. ACK1 and BRK non-receptor tyrosine kinase deficiencies are associated with familial systemic lupus and involved in efferocytosis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.15.24302255. [PMID: 38883731 PMCID: PMC11177913 DOI: 10.1101/2024.02.15.24302255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Systemic Lupus Erythematosus (SLE) is an autoimmune disease, the pathophysiology and genetic basis of which are incompletely understood. Using a forward genetic screen in multiplex families with systemic lupus erythematosus (SLE) we identified an association between SLE and compound heterozygous deleterious variants in the non-receptor tyrosine kinases (NRTKs) ACK1 and BRK. Experimental blockade of ACK1 or BRK increased circulating autoantibodies in vivo in mice and exacerbated glomerular IgG deposits in an SLE mouse model. Mechanistically, non-receptor tyrosine kinases (NRTKs) regulate activation, migration, and proliferation of immune cells. We found that the patients' ACK1 and BRK variants impair efferocytosis, the MERTK-mediated anti-inflammatory response to apoptotic cells, in human induced Pluripotent Stem Cell (hiPSC)-derived macrophages, which may contribute to SLE pathogenesis. Overall, our data suggest that ACK1 and BRK deficiencies are associated with human SLE and impair efferocytosis in macrophages.
Collapse
Affiliation(s)
- Stephanie Guillet
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Ecole doctorale Bio Sorbonne Paris Cité, Université Paris Descartes-Sorbonne Paris Cité.Paris, France
| | - Tomi Lazarov
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of MedicalSciences, New York, New York 10065, USA
| | - Natasha Jordan
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), King’s College London and Louise Coote Lupus Unit, Guy’s and Thomas’ Hospitals, London SE1 1UL, UK
| | - Bertrand Boisson
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, 10065 NY, USA
- University of Paris Cité, Imagine Institute, Paris, France
| | - Maria Tello
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Barbara Craddock
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY, 11794-8661
| | - Ting Zhou
- SKI Stem Cell Research Core, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Chihiro Nishi
- Laboratory of Biochemistry & Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871
| | - Rohan Bareja
- Cary and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Meyer Cancer Center Weill Cornell Medical College, New York, New York 10065, USA
| | - Hairu Yang
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | | | | | - Sabrina D. Dyall
- Department of Biosciences and Ocean Studies, Faculty of Science, University of Mauritius, Reduit, Mauritius
| | - David Isenberg
- Centre for Rheumatology, Division of Medicine, University College London, The Rayne Building, University College London
| | - David D’Cruz
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), King’s College London and Louise Coote Lupus Unit, Guy’s and Thomas’ Hospitals, London SE1 1UL, UK
| | - Nico Lachmann
- Institute of Experimental Hematology, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover 30625, Germany
| | - Olivier Elemento
- Cary and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Meyer Cancer Center Weill Cornell Medical College, New York, New York 10065, USA
| | - Agnes Viale
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Nicholas D. Socci
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Bioinformatics Core, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Laurent Abel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, 10065 NY, USA
- University of Paris Cité, Imagine Institute, Paris, France
| | - Shigekazu Nagata
- Laboratory of Biochemistry & Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871
| | - Morgan Huse
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - W. Todd Miller
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY, 11794-8661
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, 10065 NY, USA
- University of Paris Cité, Imagine Institute, Paris, France
- Howard Hughes Medical Institute, New York, 10065 NY, USA
- Lab of Human Genetics of Infectious Diseases, INSERM, Necker Hospital for Sick Children, Paris, France, EU
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, France, EU
| | - Frederic Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of MedicalSciences, New York, New York 10065, USA
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), King’s College London and Louise Coote Lupus Unit, Guy’s and Thomas’ Hospitals, London SE1 1UL, UK
| |
Collapse
|