1
|
Sarkar P, Wang X, Hu W, Zhu J, Ho WZ. Human Microglia Models for NeuroHIV. Viruses 2025; 17:641. [PMID: 40431653 PMCID: PMC12116007 DOI: 10.3390/v17050641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/14/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
Microglia are the primary target and reservoir of HIV infection in the central nervous system (CNS), which contributes to HIV-associated neurocognitive disorder (HAND). However, studying HIV infection of microglia has been challenged by the limited availability of primary human microglial cells. To overcome this issue, investigators have developed various microglial models for HIV studies, including immortalized human microglial cell lines, HIV latently infected microglial clones, peripheral blood monocyte-derived microglia (MMG), induced pluripotent stem cell (iPSC)-derived microglia (iMg), and microglia-containing cerebral organoids (MCOs) from iPSCs. Though these models have been used in many laboratories, the published data about their expression of the specific human microglia markers and the HIV entry receptors are conflicting. In addition, there is limited information about their feasibility and applicability as a suitable model for acute and/or latent HIV infection. This review provides a concise summary of the currently used human microglial models, with a focus on their suitability for NeuroHIV research.
Collapse
Affiliation(s)
- Priyanka Sarkar
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA; (P.S.); (X.W.)
| | - Xu Wang
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA; (P.S.); (X.W.)
| | - Wenhui Hu
- Department of Neuroscience, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Jian Zhu
- Department of Pathology, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA;
| | - Wen-Zhe Ho
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA; (P.S.); (X.W.)
| |
Collapse
|
2
|
Hemati H, Blanton MB, True HE, Koura J, Khadka R, Grant KA, Messaoudi I. Phenotypic and Functional Alterations in Peripheral Blood Mononuclear Cell-Derived Microglia in a Primate Model of Chronic Alcohol Consumption. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636708. [PMID: 39975066 PMCID: PMC11839131 DOI: 10.1101/2025.02.05.636708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Alcohol-induced dysregulation of microglial activity is associated with neuroinflammation, cognitive decline, heightened risk for neurodegenerative diseases, alcohol dependence, and escalation of alcohol drinking. Given the challenge of longitudinally sampling primary microglia, we optimized an in vitro method to differentiate peripheral blood mononuclear cells (PBMC) from non-human primates (NHP) into microglia-like cells (induced-microglia; iMGL). The iMGLs displayed transcriptional profiles distinct from those of monocyte progenitors and closely resembling those of primary microglia. Notably, morphological features showed that differentiated iMGLs derived from NHPs with chronic alcohol consumption (CAC) possessed a more mature-like microglial morphology. Additionally, dysregulation in key inflammatory and regulatory markers alongside increased baseline phagocytic activity was observed in CAC-derived IMGLs in the resting state. Phenotypic and functional assessments following LPS stimulation indicated the presence of an immune-tolerant phenotype and enrichment of a CD86+ hyper-inflammatory subpopulation in iMGLs derived from ethanol-exposed animals. Collectively, these findings demonstrate that in vitro differentiation of PBMC offers a minimally invasive approach to studying the impact of CAC on microglial function revealing that CAC reshapes both functional and transcriptional profiles of microglia.
Collapse
Affiliation(s)
- Hami Hemati
- Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Madison B Blanton
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Heather E True
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Jude Koura
- Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Rupak Khadka
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, OR, United States
| | - Kathleen A Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, OR, United States
| | - Ilhem Messaoudi
- Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
3
|
Schepanski S, Ngoumou GB, Buss C, Seifert G. Assessing in-vitro models for microglial development and fetal programming: a critical review. Front Immunol 2025; 16:1538920. [PMID: 39944696 PMCID: PMC11814449 DOI: 10.3389/fimmu.2025.1538920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/08/2025] [Indexed: 05/09/2025] Open
Abstract
This review evaluates in-vitro models for studying how maternal influences during pregnancy impact the development of offspring microglia, the immune cells of the central nervous system. The models examined include primary microglia cultures, microglia cell lines, iPSC-derived microglia, PBMC-induced microglia-like cells, 3D brain organoids derived from iPSCs, and Hofbauer cells. Each model is assessed for its ability to replicate the in-vivo environment of the developing brain, with a focus on their strengths, limitations, and practical challenges. Key factors such as scalability, genetic and epigenetic fidelity, and physiological relevance are highlighted. Microglia cell lines are highly scalable but lack genetic and epigenetic fidelity. iPSC-derived microglia provide moderate physiological relevance and patient-specific genetic insights but face operational and epigenetic challenges inherent to reprogramming. 3D brain organoids, derived from iPSCs, offer an advanced platform for studying complex neurodevelopmental processes but require extensive resources and technical expertise. Hofbauer cells, which are fetal macrophages located in the placenta and share a common developmental origin with microglia, are uniquely exposed to prenatal maternal factors and, depending on fetal barrier maturation, exhibit variable epigenetic fidelity. This makes them particularly useful for exploring the impact of maternal influences on fetal programming of microglial development. The review concludes that no single model comprehensively captures all aspects of maternal influences on microglial development, but it offers guidance on selecting the most appropriate model based on specific research objectives and experimental constraints.
Collapse
Affiliation(s)
- Steven Schepanski
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité Competence Center for Traditional and Integrative Medicine (CCCTIM), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Pediatrics, Division of Oncology and Hematology, Berlin, Germany
| | - Gonza B. Ngoumou
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité Competence Center for Traditional and Integrative Medicine (CCCTIM), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Pediatrics, Division of Oncology and Hematology, Berlin, Germany
| | - Claudia Buss
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Medical Psychology, Berlin, Germany
- University of California, Irvine, Development, Health and Disease Research Program, Irvine, CA, United States
- German Center for Child and Adolescent Health (DZKJ), Partner Site Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Mental Health (DZPG), Partner Site Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Georg Seifert
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité Competence Center for Traditional and Integrative Medicine (CCCTIM), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Pediatrics, Division of Oncology and Hematology, Berlin, Germany
| |
Collapse
|
4
|
Doratt BM, True HE, Sureshchandra S, Qiao Q, Rincon M, Marshall NE, Messaoudi I. The immune landscape of fetal chorionic villous tissue in term placenta. Front Immunol 2025; 15:1506305. [PMID: 39872537 PMCID: PMC11769816 DOI: 10.3389/fimmu.2024.1506305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/18/2024] [Indexed: 01/30/2025] Open
Abstract
Introduction The immune compartment within fetal chorionic villi is comprised of fetal Hofbauer cells (HBC) and invading placenta-associated maternal monocytes and macrophages (PAMM). Recent studies have characterized the transcriptional profile of the first trimester (T1) placenta; however, the phenotypic and functional diversity of chorionic villous immune cells at term (T3) remain poorly understood. Methods To address this knowledge gap, immune cells from human chorionic villous tissues obtained from full-term, uncomplicated pregnancies were deeply phenotyped using a combination of flow cytometry, single-cell RNA sequencing (scRNA-seq, CITE-seq) and chromatin accessibility profiling (snATAC-seq). Results Our results indicate that, relative to the first trimester, the frequency of fetal macrophages (HBC, proliferating HBC) is significantly reduced, whereas that of infiltrating maternal monocytes/macrophages (PAMM1b, PAMM1a, PAMM2, MAC_1) increased in T3. PAMM1b and HBCs exhibit the most phagocytic capacity at term highlighting their regulatory role in tissue homeostasis in late pregnancy. The transcriptional profiles of resident villous immune subsets exhibit a heightened activation state relative to the relative to T1, likely to support labor and parturition. Additionally, we provide one of the first insights into the chromatin accessibility profile of villous myeloid cells at term. We next stratified our findings by pre-pregnancy BMI since maternal pregravid obesity is associated with several adverse pregnancy outcomes. Pregravid obesity increased inflammatory gene expression, particularly among HBC and PAMM1a subsets, but dampened the expression of antimicrobial genes, supporting a tolerant-like phenotype of chorionic villous myeloid cells. We report a decline in HBC abundance accompanied by an increase in infiltrating maternal macrophages, which aligns with reports of heightened chorionic villous inflammatory pathologies with pregravid obesity. Finally, given the shared fetal yolk-sac origin of HBCs and microglia, we leveraged an in vitro model of umbilical cord blood-derived microglia to investigate the impact of pregravid obesity on fetal neurodevelopment. Our findings reveal increased expression of activation markers albeit dampened phagocytic capacity in microglia with pregravid obesity. Discussion Overall, our study highlights immune adaptations in the fetal chorionic villous with gestational age and pregravid obesity, as well as insight towards microglia dysfunction possibly underlying poor neurodevelopmental outcomes in offspring of women with pregravid obesity.
Collapse
Affiliation(s)
- Brianna M. Doratt
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, United States
| | - Heather E. True
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, United States
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, United States
| | - Suhas Sureshchandra
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
- Institute for Immunology, University of California, Irvine, Irvine, CA, United States
| | - Qi Qiao
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, United States
- Department of Biostatistics, University of Kentucky, Lexington, KY, United States
| | - Monica Rincon
- Maternal-Fetal Medicine, Oregon Health and Science University, Portland, OR, United States
| | - Nicole E. Marshall
- Maternal-Fetal Medicine, Oregon Health and Science University, Portland, OR, United States
| | - Ilhem Messaoudi
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
5
|
Shook LL, Batorsky RE, De Guzman RM, McCrea LT, Brigida SM, Horng JE, Sheridan SD, Kholod O, Cook AM, Li JZ, Slonim DK, Goods BA, Perlis RH, Edlow AG. Maternal SARS-CoV-2 impacts fetal placental macrophage programs and placenta-derived microglial models of neurodevelopment. J Neuroinflammation 2024; 21:163. [PMID: 38918792 PMCID: PMC11197235 DOI: 10.1186/s12974-024-03157-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND The SARS-CoV-2 virus activates maternal and placental immune responses. Such activation in the setting of other infections during pregnancy is known to impact fetal brain development. The effects of maternal immune activation on neurodevelopment are mediated at least in part by fetal brain microglia. However, microglia are inaccessible for direct analysis, and there are no validated non-invasive surrogate models to evaluate in utero microglial priming and function. We have previously demonstrated shared transcriptional programs between microglia and Hofbauer cells (HBCs, or fetal placental macrophages) in mouse models. METHODS AND RESULTS We assessed the impact of maternal SARS-CoV-2 on HBCs isolated from 24 term placentas (N = 10 SARS-CoV-2 positive cases, 14 negative controls). Using single-cell RNA-sequencing, we demonstrated that HBC subpopulations exhibit distinct cellular programs, with specific subpopulations differentially impacted by SARS-CoV-2. Assessment of differentially expressed genes implied impaired phagocytosis, a key function of both HBCs and microglia, in some subclusters. Leveraging previously validated models of microglial synaptic pruning, we showed that HBCs isolated from placentas of SARS-CoV-2 positive pregnancies can be transdifferentiated into microglia-like cells (HBC-iMGs), with impaired synaptic pruning behavior compared to HBC models from negative controls. CONCLUSION These findings suggest that HBCs isolated at birth can be used to create personalized cellular models of offspring microglial programming.
Collapse
Affiliation(s)
- Lydia L Shook
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, 55 Fruit Street, Thier Research Building, 903B, Boston, MA, 02114, USA
- Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, MA, USA
| | | | - Rose M De Guzman
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, 55 Fruit Street, Thier Research Building, 903B, Boston, MA, 02114, USA
- Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Liam T McCrea
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sara M Brigida
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, 55 Fruit Street, Thier Research Building, 903B, Boston, MA, 02114, USA
| | - Joy E Horng
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Steven D Sheridan
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Olha Kholod
- Thayer School of Engineering and Program, Dartmouth College, Hanover, NH, USA
| | - Aidan M Cook
- Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Jonathan Z Li
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Donna K Slonim
- Department of Computer Science, Tufts University, Medford, MA, USA
| | - Brittany A Goods
- Thayer School of Engineering and Program, Dartmouth College, Hanover, NH, USA
- Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Roy H Perlis
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrea G Edlow
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, 55 Fruit Street, Thier Research Building, 903B, Boston, MA, 02114, USA.
- Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Sheridan SD, Horng JE, Yeh H, McCrea L, Wang J, Fu T, Perlis RH. Loss of Function in the Neurodevelopmental Disease and Schizophrenia-Associated Gene CYFIP1 in Human Microglia-like Cells Supports a Functional Role in Synaptic Engulfment. Biol Psychiatry 2024; 95:676-686. [PMID: 37573007 PMCID: PMC10874584 DOI: 10.1016/j.biopsych.2023.07.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 07/18/2023] [Accepted: 07/23/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND The CYFIP1 gene, located in the neurodevelopmental risk locus 15q11.2, is highly expressed in microglia, but its role in human microglial function as it relates to neurodevelopment is not well understood. METHODS We generated multiple CRISPR (clustered regularly interspaced short palindromic repeat) knockouts of CYFIP1 in patient-derived models of microglia to characterize function and phenotype. Using microglia-like cells reprogrammed from peripheral blood mononuclear cells, we quantified phagocytosis of synaptosomes (isolated and purified synaptic vesicles) from human induced pluripotent stem cell (iPSC)-derived neuronal cultures as an in vitro model of synaptic pruning. We repeated these analyses in human iPSC-derived microglia-like cells derived from 3 isogenic wild-type/knockout line pairs derived from 2 donors and further characterized microglial development and function through morphology and motility. RESULTS CYFIP1 knockout using orthogonal CRISPR constructs in multiple patient-derived cell lines was associated with a statistically significant decrease in synaptic vesicle phagocytosis in microglia-like cell models derived from both peripheral blood mononuclear cells and iPSCs. Morphology was also shifted toward a more ramified profile, and motility was significantly reduced. However, iPSC-CYFIP1 knockout lines retained the ability to differentiate to functional microglia. CONCLUSIONS The changes in microglial phenotype and function due to the loss of function of CYFIP1 observed in this study implicate a potential impact on processes such as synaptic pruning that may contribute to CYFIP1-related neurodevelopmental disorders. Investigating risk genes in a range of central nervous system cell types, not solely neurons, may be required to fully understand the way in which common and rare variants intersect to yield neuropsychiatric disorders.
Collapse
Affiliation(s)
- Steven D Sheridan
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Joy E Horng
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Hana Yeh
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Liam McCrea
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Jennifer Wang
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Ting Fu
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Roy H Perlis
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
7
|
Chagas LDS, Serfaty CA. The Influence of Microglia on Neuroplasticity and Long-Term Cognitive Sequelae in Long COVID: Impacts on Brain Development and Beyond. Int J Mol Sci 2024; 25:3819. [PMID: 38612629 PMCID: PMC11011312 DOI: 10.3390/ijms25073819] [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: 03/01/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Microglial cells, the immune cells of the central nervous system, are key elements regulating brain development and brain health. These cells are fully responsive to stressors, microenvironmental alterations and are actively involved in the construction of neural circuits in children and the ability to undergo full experience-dependent plasticity in adults. Since neuroinflammation is a known key element in the pathogenesis of COVID-19, one might expect the dysregulation of microglial function to severely impact both functional and structural plasticity, leading to the cognitive sequelae that appear in the pathogenesis of Long COVID. Therefore, understanding this complex scenario is mandatory for establishing the possible molecular mechanisms related to these symptoms. In the present review, we will discuss Long COVID and its association with reduced levels of BDNF, altered crosstalk between circulating immune cells and microglia, increased levels of inflammasomes, cytokines and chemokines, as well as the alterations in signaling pathways that impact neural synaptic remodeling and plasticity, such as fractalkines, the complement system, the expression of SIRPα and CD47 molecules and altered matrix remodeling. Together, these complex mechanisms may help us understand consequences of Long COVID for brain development and its association with altered brain plasticity, impacting learning disabilities, neurodevelopmental disorders, as well as cognitive decline in adults.
Collapse
Affiliation(s)
- Luana da Silva Chagas
- Program of Neuroscience, Department of Neurobiology, Institute of Biology, Federal Fluminense University, Niterói 24210-201, Rio de Janeiro, Brazil;
- National Institute of Science and Technology on Neuroimmunomodulation—INCT-NIM, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, Rio de Janeiro, Brazil
- Rio de Janeiro Research Network on Neuroinflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, Rio de Janeiro, Brazil
| | - Claudio Alberto Serfaty
- Program of Neuroscience, Department of Neurobiology, Institute of Biology, Federal Fluminense University, Niterói 24210-201, Rio de Janeiro, Brazil;
- National Institute of Science and Technology on Neuroimmunomodulation—INCT-NIM, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, Rio de Janeiro, Brazil
- Rio de Janeiro Research Network on Neuroinflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, Rio de Janeiro, Brazil
| |
Collapse
|
8
|
Jäntti H, Kistemaker L, Buonfiglioli A, De Witte LD, Malm T, Hol EM. Emerging Models to Study Human Microglia In vitro. ADVANCES IN NEUROBIOLOGY 2024; 37:545-568. [PMID: 39207712 DOI: 10.1007/978-3-031-55529-9_30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
New in vitro models provide an exciting opportunity to study live human microglia. Previously, a major limitation in understanding human microglia in health and disease has been their limited availability. Here, we provide an overview of methods to obtain human stem cell or blood monocyte-derived microglia-like cells that provide a nearly unlimited source of live human microglia for research. We address how understanding microglial ontogeny can help modeling microglial identity and function in a dish with increased accuracy. Moreover, we categorize stem cell-derived differentiation methods into embryoid body based, growth factor driven, and coculture-driven approaches, and review novel viral approaches to reprogram stem cells directly into microglia-like cells. Furthermore, we review typical readouts used in the field to verify microglial identity and characterize functional microglial phenotypes. We provide an overview of methods used to study microglia in environments more closely resembling the (developing) human CNS, such as cocultures and brain organoid systems with incorporated or innately developing microglia. We highlight how microglia-like cells can be utilized to reveal molecular and functional mechanisms in human disease context, focusing on Alzheimer's disease and other neurodegenerative diseases as well as neurodevelopmental diseases. Finally, we provide a critical overview of challenges and future opportunities to more accurately model human microglia in a dish and conclude that novel in vitro microglia-like cells provide an exciting potential to bring preclinical research of microglia to a new era.
Collapse
Affiliation(s)
- Henna Jäntti
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Lois Kistemaker
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Alice Buonfiglioli
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lot D De Witte
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Elly M Hol
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
9
|
Shook LL, Batorsky RA, De Guzman RM, McCrea LT, Brigida SM, Horng JE, Sheridan SD, Kholod O, Cook AM, Li JZ, Goods BA, Perlis RH, Edlow AG. Maternal SARS-CoV-2 impacts fetal placental macrophage programs and placenta-derived microglial models of neurodevelopment. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.29.23300544. [PMID: 38234776 PMCID: PMC10793528 DOI: 10.1101/2023.12.29.23300544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
The SARS-CoV-2 virus activates maternal and placental immune responses, which in the setting of other infections occurring during pregnancy are known to impact fetal brain development. The effects of maternal immune activation on neurodevelopment are mediated at least in part by fetal brain microglia. However, microglia are inaccessible for direct analysis, and there are no validated non-invasive surrogate models to evaluate in utero microglial priming and function. We have previously demonstrated shared transcriptional programs between microglia and Hofbauer cells (HBCs, or fetal placental macrophages) in mouse models. Here, we assessed the impact of maternal SARS-CoV-2 on HBCs isolated from term placentas using single-cell RNA-sequencing. We demonstrated that HBC subpopulations exhibit distinct cellular programs, with specific subpopulations differentially impacted by SARS-CoV-2. Assessment of differentially expressed genes implied impaired phagocytosis, a key function of both HBCs and microglia, in some subclusters. Leveraging previously validated models of microglial synaptic pruning, we showed that HBCs isolated from placentas of SARS-CoV-2 positive pregnancies can be transdifferentiated into microglia-like cells, with altered morphology and impaired synaptic pruning behavior compared to HBC models from negative controls. These findings suggest that HBCs isolated at birth can be used to create personalized cellular models of offspring microglial programming.
Collapse
|
10
|
McMillan RE, Wang E, Carlin AF, Coufal NG. Human microglial models to study host-virus interactions. Exp Neurol 2023; 363:114375. [PMID: 36907350 PMCID: PMC10521930 DOI: 10.1016/j.expneurol.2023.114375] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/13/2023] [Accepted: 03/02/2023] [Indexed: 03/14/2023]
Abstract
Microglia, the resident macrophage of the central nervous system, are increasingly recognized as contributing to diverse aspects of human development, health, and disease. In recent years, numerous studies in both mouse and human models have identified microglia as a "double edged sword" in the progression of neurotropic viral infections: protecting against viral replication and cell death in some contexts, while acting as viral reservoirs and promoting excess cellular stress and cytotoxicity in others. It is imperative to understand the diversity of human microglial responses in order to therapeutically modulate them; however, modeling human microglia has been historically challenging due to significant interspecies differences in innate immunity and rapid transformation upon in vitro culture. In this review, we discuss the contribution of microglia to the neuropathogenesis of key neurotropic viral infections: human immunodeficiency virus 1 (HIV-1), Zika virus (ZIKV), Japanese encephalitis virus (JEV), West Nile virus (WNV), Herpes simplex virus (HSV), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We pay special attention to recent work with human stem cell-derived microglia and propose strategies to leverage these powerful models to further uncover species- and disease-specific microglial responses and novel therapeutic interventions for neurotropic viral infections.
Collapse
Affiliation(s)
- Rachel E McMillan
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, United States of America; Department of Pathology and Medicine, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States of America
| | - Ellen Wang
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States of America; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92093, United States of America
| | - Aaron F Carlin
- Department of Pathology and Medicine, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States of America.
| | - Nicole G Coufal
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States of America; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92093, United States of America.
| |
Collapse
|
11
|
Alqahtani MS, Abbas M, Alshahrani MY, Alabdullh K, Alqarni A, Alqahtani FF, Jambi LK, Alkhayat A. Effects of COVID-19 on Synaptic and Neuronal Degeneration. Brain Sci 2023; 13:brainsci13010131. [PMID: 36672112 PMCID: PMC9856402 DOI: 10.3390/brainsci13010131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/31/2022] [Accepted: 01/04/2023] [Indexed: 01/13/2023] Open
Abstract
Neurons are the basic building blocks of the human body's neurological system. Atrophy is defined by the disintegration of the connections between cells that enable them to communicate. Peripheral neuropathy and demyelinating disorders, as well as cerebrovascular illnesses and central nervous system (CNS) inflammatory diseases, have all been linked to brain damage, including Parkinson's disease (PD). It turns out that these diseases have a direct impact on brain atrophy. However, it may take some time after the onset of one of these diseases for this atrophy to be clearly diagnosed. With the emergence of the Coronavirus disease 2019 (COVID-19) pandemic, there were several clinical observations of COVID-19 patients. Among those observations is that the virus can cause any of the diseases that can lead to brain atrophy. Here we shed light on the research that tracked the relationship of these diseases to the COVID-19 virus. The importance of this review is that it is the first to link the relationship between the Coronavirus and diseases that cause brain atrophy. It also indicates the indirect role of the virus in dystrophy.
Collapse
Affiliation(s)
- Mohammed S. Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia
- BioImaging Unit, Space Research Centre, University of Leicester, Michael Atiyah Building, Leicester LE1 7RH, UK
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia
- Electronics and Communications Department, College of Engineering, Delta University for Science and Technology, Gamasa 35712, Egypt
- Correspondence:
| | - Mohammad Y. Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia
| | - Khulud Alabdullh
- Radiology Department, King Abdullah Hospital Bisha, Bisha 61922, Saudi Arabia
| | - Amjad Alqarni
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia
| | - Fawaz F. Alqahtani
- Department of Radiological Sciences, College of Applied Medical Sciences, Najran University, Najran 55461, Saudi Arabia
| | - Layal K. Jambi
- Radiological Sciences Department, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia
| | - Adnan Alkhayat
- Department of Hematopathology, King Fahad Central Hospital, Gizan 82666, Saudi Arabia
| |
Collapse
|
12
|
Sargeant TJ, Fourrier C. Human monocyte-derived microglia-like cell models: A review of the benefits, limitations and recommendations. Brain Behav Immun 2023; 107:98-109. [PMID: 36202170 DOI: 10.1016/j.bbi.2022.09.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 09/09/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022] Open
Abstract
In the last few decades, mounting evidence has highlighted that microglia have crucial roles in both health and disease. This has led to a growing interest in studying human microglia in disease-relevant models. However, current models present limitations that can make them unsuitable for moderate throughput studies in human cohorts. Primary human microglia are ethically and technically difficult to obtain and only allow low throughput studies; immortalized cell lines have been shown to differ too greatly from primary human microglia; and induced pluripotent stem cell-derived microglia, although physiologically relevant in most contexts, have limited potential for use in large cohorts of people or for personalised drug screening. In this review, we discuss monocyte-derived microglia-like (MDMi) cells, a model that has been developed and increasingly used in the last decade, using human monocytes isolated from blood samples. We describe the variety of protocols that have been used to develop MDMi cell models and highlight a need for standardization across protocols. We then summarize data that validate MDMi cells as an appropriate model to study human microglia in health and disease. We also present the benefits and limitations of using this approach to study human microglia compared with other microglial models, when used in combination with the relevant downstream applications and with cross-validation of findings in other systems. Finally, we summarize the paradigms in which MDMi models have been used to advance research on microglia in immune-related disease. This review is an important reference for scientists who seek to establish MDMi cells as a microglial model for the advancement of our understanding of microglia in human health and disease.
Collapse
Affiliation(s)
- Timothy J Sargeant
- Lysosomal Health in Ageing, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia.
| | - Célia Fourrier
- Lysosomal Health in Ageing, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
13
|
Shook LL, Fourman LT, Edlow AG. Immune Responses to SARS-CoV-2 in Pregnancy: Implications for the Health of the Next Generation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:1465-1473. [PMID: 36192115 PMCID: PMC9536183 DOI: 10.4049/jimmunol.2200414] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/08/2022] [Indexed: 11/06/2022]
Abstract
Widespread SARS-CoV-2 infection among pregnant individuals has led to a generation of fetuses exposed in utero, but the long-term impact of such exposure remains unknown. Although fetal infection is rare, children born to mothers with SARS-CoV-2 infection may be at increased risk for adverse neurodevelopmental and cardiometabolic outcomes. Fetal programming effects are likely to be mediated at least in part by maternal immune activation. In this review, we discuss recent evidence regarding the effects of prenatal SARS-CoV-2 infection on the maternal, placental, and fetal immune response, as well as the implications for the long-term health of offspring. Extrapolating from what is known about the impact of maternal immune activation in other contexts (e.g., obesity, HIV, influenza), we review the potential for neurodevelopmental and cardiometabolic morbidity in offspring. Based on available data suggesting potential increased neurodevelopmental risk, we highlight the importance of establishing large cohorts to monitor offspring born to SARS-CoV-2-positive mothers for neurodevelopmental and cardiometabolic sequelae.
Collapse
Affiliation(s)
- Lydia L Shook
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA; and
| | - Lindsay T Fourman
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Andrea G Edlow
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Massachusetts General Hospital and Harvard Medical School, Boston, MA;
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA; and
| |
Collapse
|
14
|
Sheridan SD, Horng JE, Perlis RH. Patient-Derived In Vitro Models of Microglial Function and Synaptic Engulfment in Schizophrenia. Biol Psychiatry 2022; 92:470-479. [PMID: 35232567 PMCID: PMC10039432 DOI: 10.1016/j.biopsych.2022.01.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/19/2021] [Accepted: 01/10/2022] [Indexed: 01/11/2023]
Abstract
Multiple lines of evidence implicate dysregulated microglia-mediated synaptic pruning in the pathophysiology of schizophrenia. In vitro human cellular studies represent a promising means of pursuing this hypothesis, complementing efforts with animal models and postmortem human data while addressing their limitations. The challenges in culturing homogeneous populations of cells derived from postmortem or surgical biopsy brain material from patients, and their limited availability, has led to a focus on differentiation of induced pluripotent stem cells. These methods too have limitations, in that they disrupt the epigenome and can demonstrate line-to-line variability due in part to extended time in culture, partial reprogramming, and/or residual epigenetic memory from the cell source, yielding large technical artifacts. Yet another strategy uses direct transdifferentiation of peripheral mononuclear blood cells, or umbilical cord blood cells, to microglia-like cells. Any of these approaches can be paired with patient-derived synaptosomes from differentiated neurons as a simpler alternative to co-culture. Patient-derived microglia models may facilitate identification of novel modulators of synaptic pruning and identification of biomarkers that may allow more targeted early interventions.
Collapse
Affiliation(s)
- Steven D Sheridan
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Joy E Horng
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Roy H Perlis
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
15
|
Cuní-López C, Stewart R, Quek H, White AR. Recent Advances in Microglia Modelling to Address Translational Outcomes in Neurodegenerative Diseases. Cells 2022; 11:cells11101662. [PMID: 35626698 PMCID: PMC9140031 DOI: 10.3390/cells11101662] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases are deteriorating conditions of the nervous system that are rapidly increasing in the aging population. Increasing evidence suggests that neuroinflammation, largely mediated by microglia, the resident immune cells of the brain, contributes to the onset and progression of neurodegenerative diseases. Hence, microglia are considered a major therapeutic target that could potentially yield effective disease-modifying treatments for neurodegenerative diseases. Despite the interest in studying microglia as drug targets, the availability of cost-effective, flexible, and patient-specific microglia cellular models is limited. Importantly, the current model systems do not accurately recapitulate important pathological features or disease processes, leading to the failure of many therapeutic drugs. Here, we review the key roles of microglia in neurodegenerative diseases and provide an update on the current microglia platforms utilised in neurodegenerative diseases, with a focus on human microglia-like cells derived from peripheral blood mononuclear cells as well as human-induced pluripotent stem cells. The described microglial platforms can serve as tools for investigating disease biomarkers and improving the clinical translatability of the drug development process in neurodegenerative diseases.
Collapse
Affiliation(s)
- Carla Cuní-López
- Cell & Molecular Biology Department, Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (C.C.-L.); (R.S.)
- Faculty of Medicine, The University of Queensland, Brisbane, QLD 4006, Australia
| | - Romal Stewart
- Cell & Molecular Biology Department, Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (C.C.-L.); (R.S.)
- UQ Centre for Clinical Research, The University of Queensland, Royal Brisbane & Women’s Hospital, Brisbane, QLD 4006, Australia
| | - Hazel Quek
- Cell & Molecular Biology Department, Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (C.C.-L.); (R.S.)
- Correspondence: (H.Q.); (A.R.W.)
| | - Anthony R. White
- Cell & Molecular Biology Department, Mental Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (C.C.-L.); (R.S.)
- Correspondence: (H.Q.); (A.R.W.)
| |
Collapse
|
16
|
Shook LL, Sullivan EL, Lo JO, Perlis RH, Edlow AG. COVID-19 in pregnancy: implications for fetal brain development. Trends Mol Med 2022; 28:319-330. [PMID: 35277325 PMCID: PMC8841149 DOI: 10.1016/j.molmed.2022.02.004] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 02/08/2022] [Accepted: 02/08/2022] [Indexed: 11/02/2022]
Abstract
The impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection during pregnancy on the developing fetal brain is poorly understood. Other antenatal infections such as influenza have been associated with adverse neurodevelopmental outcomes in offspring. Although vertical transmission has been rarely observed in SARS-CoV-2 to date, given the potential for profound maternal immune activation (MIA), impact on the developing fetal brain is likely. Here we review evidence that SARS-CoV-2 and other viral infections during pregnancy can result in maternal, placental, and fetal immune activation, and ultimately in offspring neurodevelopmental morbidity. Finally, we highlight the need for cellular models of fetal brain development to better understand potential short- and long-term impacts of maternal SARS-CoV-2 infection on the next generation.
Collapse
Affiliation(s)
- Lydia L Shook
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Elinor L Sullivan
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, USA; Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA; Division of Neuroscience, Oregon National Primate Center, Beaverton, OR, USA
| | - Jamie O Lo
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, OR, USA; Department of Urology, Oregon Health & Science University, Portland, OR, USA; Division of Reproductive and Developmental Sciences, Oregon National Primate Center, Beaverton, OR, USA
| | - Roy H Perlis
- Center for Quantitative Health, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrea G Edlow
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
17
|
Quek H, Cuní-López C, Stewart R, Colletti T, Notaro A, Nguyen TH, Sun Y, Guo CC, Lupton MK, Roberts TL, Lim YC, Oikari LE, La Bella V, White AR. ALS monocyte-derived microglia-like cells reveal cytoplasmic TDP-43 accumulation, DNA damage, and cell-specific impairment of phagocytosis associated with disease progression. J Neuroinflammation 2022; 19:58. [PMID: 35227277 PMCID: PMC8887023 DOI: 10.1186/s12974-022-02421-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 02/21/2022] [Indexed: 02/06/2023] Open
Abstract
Abstract
Background
Amyotrophic lateral sclerosis (ALS) is a multifactorial neurodegenerative disease characterised by the loss of upper and lower motor neurons. Increasing evidence indicates that neuroinflammation mediated by microglia contributes to ALS pathogenesis. This microglial activation is evident in post-mortem brain tissues and neuroimaging data from patients with ALS. However, the role of microglia in the pathogenesis and progression of amyotrophic lateral sclerosis remains unclear, partly due to the lack of a model system that is able to faithfully recapitulate the clinical pathology of ALS. To address this shortcoming, we describe an approach that generates monocyte-derived microglia-like cells that are capable of expressing molecular markers, and functional characteristics similar to in vivo human brain microglia.
Methods
In this study, we have established monocyte-derived microglia-like cells from 30 sporadic patients with ALS, including 15 patients with slow disease progression, 6 with intermediate progression, and 9 with rapid progression, together with 20 non-affected healthy controls.
Results
We demonstrate that patient monocyte-derived microglia-like cells recapitulate canonical pathological features of ALS including non-phosphorylated and phosphorylated-TDP-43-positive inclusions. Moreover, ALS microglia-like cells showed significantly impaired phagocytosis, altered cytokine profiles, and abnormal morphologies consistent with a neuroinflammatory phenotype. Interestingly, all ALS microglia-like cells showed abnormal phagocytosis consistent with the progression of the disease. In-depth analysis of ALS microglia-like cells from the rapid disease progression cohort revealed significantly altered cell-specific variation in phagocytic function. In addition, DNA damage and NOD-leucine rich repeat and pyrin containing protein 3 (NLRP3) inflammasome activity were also elevated in ALS patient monocyte-derived microglia-like cells, indicating a potential new pathway involved in driving disease progression.
Conclusions
Taken together, our work demonstrates that the monocyte-derived microglia-like cell model recapitulates disease-specific hallmarks and characteristics that substantiate patient heterogeneity associated with disease subgroups. Thus, monocyte-derived microglia-like cells are highly applicable to monitor disease progression and can be applied as a functional readout in clinical trials for anti-neuroinflammatory agents, providing a basis for personalised treatment for patients with ALS.
Collapse
|
18
|
O'Connor TG, Ciesla AA. Maternal Immune Activation Hypotheses for Human Neurodevelopment: Some Outstanding Questions. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2021; 7:471-479. [PMID: 34688920 PMCID: PMC9021321 DOI: 10.1016/j.bpsc.2021.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/05/2021] [Accepted: 10/13/2021] [Indexed: 12/17/2022]
Abstract
The Maternal Immune Activation (MIA) hypothesis is a leading model for understanding prenatal influences on individual differences in, and clinical syndromes of, neurodevelopment. Experimental animal and human research has proliferated in recent years, and there is now a sizable research base. Several meta-analyses demonstrate general support for an association between prenatal immune activation and neurodevelopment in human research. However, questions remain about the nature of the immune activation, the network of underlying mechanisms involved, and the breadth of impact across behavioral phenotypes. Complementing recent reviews of results, the current review places particular emphasis on how advances in understanding mechanisms may be improved with greater attention to addressing the methodological variation and limitations of existing studies, and identifies areas for further clinical research.
Collapse
Affiliation(s)
- Thomas G O'Connor
- Department of Psychiatry, University of Rochester; Department of Psycholog, University of Rochestery; Department of Neuroscience, University of Rochester; Department of Obstetrics and Gynecology, University of Rochester; Wynne Center for Family Research, University of Rochester.
| | | |
Collapse
|