51
|
Yeh H, De Cruz MA, You Y, Ikezu S, Ikezu T. Development and characterization of in vitro inducible immortalization of a murine microglia cell line for high throughput studies. Sci Rep 2025; 15:3207. [PMID: 39863723 PMCID: PMC11762310 DOI: 10.1038/s41598-025-87543-1] [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: 10/16/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
There are few in vitro models available to study microglial physiology in a homeostatic context. Recent approaches include the human induced pluripotent stem cell model, but these can be challenging for large-scale assays and may lead to batch variability. To advance our understanding of microglial biology while enabling scalability for high-throughput assays, we developed an inducible immortalized murine microglial cell line using a tetracycline expression system. The addition of doxycycline facilitates rapid cell proliferation, allowing for population expansion. Upon withdrawal of doxycycline, this monoclonal microglial cell line differentiates, resembling in vivo microglial physiology as demonstrated by the expression of microglial genes, innate immune responses, chemotaxis, and phagocytic abilities. We utilized live imaging and various molecular techniques to functionally characterize the clonal 2E11murine microglial cell line. Transcriptomic analysis showed that the 2E11 line exhibited characteristics of immature, proliferative microglia during doxycycline induction, and further differentiation led to a more homeostatic phenotype. Treatment with transforming growth factor-β modified the transcriptome of the 2E11 cell line, affecting cellular immune pathways. Our findings indicate that the 2E11 inducible immortalized cell line is a practical and convenient tool for studying microglial biology in vitro.
Collapse
Affiliation(s)
- Hana Yeh
- Graduate Program in Neuroscience, Boston University, Boston, United States
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, United States
| | - Matthew A De Cruz
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, USA
- Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA
| | - Yang You
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, United States
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Seiko Ikezu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, United States
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Tsuneya Ikezu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, United States.
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, USA.
- Regenerative Science Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA.
- Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA.
- Department of Neuroscience , Mayo Clinic Florida , 4500 San Pablo Rd S, Jacksonville, 32224, USA, FL.
| |
Collapse
|
52
|
Penati S, Brioschi S, Cai Z, Han CZ, Colonna M. Mechanisms and environmental factors shaping the ecosystem of brain macrophages. Front Immunol 2025; 16:1539988. [PMID: 39925814 PMCID: PMC11802581 DOI: 10.3389/fimmu.2025.1539988] [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: 12/05/2024] [Accepted: 01/03/2025] [Indexed: 02/11/2025] Open
Abstract
Brain macrophages encompass two major populations: microglia in the parenchyma and border-associated macrophages (BAMs) in the extra-parenchymal compartments. These cells play crucial roles in maintaining brain homeostasis and immune surveillance. Microglia and BAMs are phenotypically and epigenetically distinct and exhibit highly specialized functions tailored to their environmental niches. Intriguingly, recent studies have shown that both microglia and BAMs originate from the same myeloid progenitor during yolk sac hematopoiesis, but their developmental fates diverge within the brain. Several works have partially unveiled the mechanisms orchestrating the development of microglia and BAMs in both mice and humans; however, many questions remain unanswered. Defining the molecular underpinnings controlling the transcriptional and epigenetic programs of microglia and BAMs is one of the upcoming challenges for the field. In this review, we outline current knowledge on ontogeny, phenotypic diversity, and the factors shaping the ecosystem of brain macrophages. We discuss insights garnered from human studies, highlighting similarities and differences compared to mice. Lastly, we address current research gaps and potential future directions in the field. Understanding how brain macrophages communicate with their local environment and how the tissue instructs their developmental trajectories and functional features is essential to fully comprehend brain physiology in homeostasis and disease.
Collapse
Affiliation(s)
- Silvia Penati
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, United States
| | - Simone Brioschi
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, United States
| | - Zhangying Cai
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, United States
| | - Claudia Z. Han
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, United States
- Brain Immunology and Glia (BIG) Center, Washington University School of Medicine in Saint Louis, Saint Louis, MO, United States
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, United States
- Brain Immunology and Glia (BIG) Center, Washington University School of Medicine in Saint Louis, Saint Louis, MO, United States
| |
Collapse
|
53
|
Gongwer MW, Etienne F, Moca EN, Chappell MS, Blagburn-Blanco SV, Riley JP, Enos AS, Haratian M, Qi A, Rojo R, Wilke SA, Pridans C, DeNardo LA, De Biase LM. Microglia regulate nucleus accumbens synaptic development and circuit function underlying threat avoidance behaviors. RESEARCH SQUARE 2025:rs.3.rs-5837701. [PMID: 39975894 PMCID: PMC11838711 DOI: 10.21203/rs.3.rs-5837701/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
While CNS microglia have well-established roles in synapse pruning during neurodevelopment, only a few studies have identified roles for microglia in synapse formation. These studies focused on the cortex and primary sensory circuits during restricted developmental time periods, leaving substantial gaps in our understanding of the early developmental functions of microglia. Here we investigated how the absence of microglia impacts synaptic development in the nucleus accumbens (NAc), a region critical for emotional regulation and motivated behaviors and where dysfunction is implicated in psychiatric disorders that arise early in life. Using a genetically modified mouse that lacks microglia (Csf1r ΔFIRE/ΔFIRE), we found blunted excitatory synapse formation in the NAc. This effect was most prominent during the second and third postnatal weeks, when we previously found microglia to be overproduced, and was accompanied by an increase in presynaptic release probability and alterations in postsynaptic kinetics. Tissue-level NAc proteomics confirmed that microglial absence impacted numerous proteins involved in synapse structure, trans-synaptic signaling, and pre-synaptic function. However, microglial absence did not perturb levels of astrocyte-derived cues and adhesive proteins that promote synaptogenesis, suggesting that reduced synapse number may be caused by absence of a microglial-derived synaptogenic cue. Although observed electrophysiological synaptic changes were largely normalized by adulthood, we identified lasting effects of microglial absence on threat avoidance behavior, and these behavioral effects were directly associated with alterations of NAc neuronal activity. Together, these results indicate a critical role for microglia in regulating the synaptic landscape of the developing NAc and in establishing functional circuits underlying adult behavioral repertoires.
Collapse
Affiliation(s)
- Michael W Gongwer
- Department of Physiology, University of California Los Angeles, CA, USA
- Neuroscience Interdepartmental Program, University of California Los Angeles, CA, USA
- UCLA Medical Scientist Training Program, University of California Los Angeles, CA, USA
| | - Fanny Etienne
- Department of Physiology, University of California Los Angeles, CA, USA
| | - Eric N Moca
- Department of Physiology, University of California Los Angeles, CA, USA
| | - Megan S Chappell
- Department of Physiology, University of California Los Angeles, CA, USA
- Neuroscience Interdepartmental Program, University of California Los Angeles, CA, USA
| | - Sara V Blagburn-Blanco
- Department of Physiology, University of California Los Angeles, CA, USA
- Neuroscience Interdepartmental Program, University of California Los Angeles, CA, USA
- UCLA Medical Scientist Training Program, University of California Los Angeles, CA, USA
| | - Jack P Riley
- Department of Physiology, University of California Los Angeles, CA, USA
| | - Alexander S Enos
- Department of Physiology, University of California Los Angeles, CA, USA
| | - Melody Haratian
- Department of Physiology, University of California Los Angeles, CA, USA
| | - Alex Qi
- Department of Psychiatry, University of California Los Angeles, CA, USA
| | - Rocio Rojo
- Institution for Regeneration and Repair, University of Edinburgh, Scotland
| | - Scott A Wilke
- Department of Psychiatry, University of California Los Angeles, CA, USA
| | - Clare Pridans
- Institution for Regeneration and Repair, University of Edinburgh, Scotland
| | - Laura A DeNardo
- Department of Physiology, University of California Los Angeles, CA, USA
| | | |
Collapse
|
54
|
Yi MH, Lee J, Moon S, So E, Bang G, Moon KS, Lee KH. Divergent Crosstalk Between Microglia and T Cells in Brain Cancers: Implications for Novel Therapeutic Strategies. Biomedicines 2025; 13:216. [PMID: 39857798 PMCID: PMC11763300 DOI: 10.3390/biomedicines13010216] [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: 12/19/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Brain cancers represent a formidable oncological challenge characterized by their aggressive nature and resistance to conventional therapeutic interventions. The tumor microenvironment has emerged as a critical determinant of tumor progression and treatment efficacy. Within this complex ecosystem, microglia and macrophages play fundamental roles, forming intricate networks with peripheral immune cell populations, particularly T cells. The precise mechanisms underlying microglial interactions with T cells and their contributions to immunosuppression remain incompletely understood. Methods: This review comprehensively examines the complex cellular dialogue between microglia and T cells in two prominent brain malignancies: primary glioblastoma and secondary brain metastases. Results: Through a comprehensive review of the current scientific literature, we explore the nuanced mechanisms through which microglial-T cell interactions modulate tumor growth and immune responses. Conclusions: Our analysis seeks to unravel the cellular communication pathways that potentially underpin tumor progression, with the ultimate goal of illuminating novel therapeutic strategies for brain cancer intervention.
Collapse
Affiliation(s)
- Min-Hee Yi
- Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Republic of Korea; (M.-H.Y.)
- Biomedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Jeollanam-do, Republic of Korea
| | - Jinkyung Lee
- Biomedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Jeollanam-do, Republic of Korea
| | - Subin Moon
- Department of Medicine, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea
| | - EunA So
- Biomedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Jeollanam-do, Republic of Korea
| | - Geonhyeok Bang
- Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Republic of Korea; (M.-H.Y.)
| | - Kyung-Sub Moon
- Department of Neurosurgery, Chonnam National University Hwasun Hospital, Hwasun 58128, Jeollanam-do, Republic of Korea;
| | - Kyung-Hwa Lee
- Biomedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Jeollanam-do, Republic of Korea
- Department of Pathology, Chonnam National University Hwasun Hospital, Hwasun 58128, Jeollanam-do, Republic of Korea
| |
Collapse
|
55
|
Gongwer MW, Etienne F, Moca EN, Chappell MS, Blagburn-Blanco SV, Riley JP, Enos AS, Haratian M, Qi A, Rojo R, Wilke SA, Pridans C, DeNardo LA, De Biase LM. Microglia regulate nucleus accumbens synaptic development and circuit function underlying threat avoidance behaviors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633068. [PMID: 39868334 PMCID: PMC11761117 DOI: 10.1101/2025.01.15.633068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
While CNS microglia have well-established roles in synapse pruning during neurodevelopment, only a few studies have identified roles for microglia in synapse formation. These studies focused on the cortex and primary sensory circuits during restricted developmental time periods, leaving substantial gaps in our understanding of the early developmental functions of microglia. Here we investigated how the absence of microglia impacts synaptic development in the nucleus accumbens (NAc), a region critical for emotional regulation and motivated behaviors and where dysfunction is implicated in psychiatric disorders that arise early in life. Using a genetically modified mouse that lacks microglia (Csf1r ΔFIRE/ΔFIRE), we found blunted excitatory synapse formation in the NAc. This effect was most prominent during the second and third postnatal weeks, when we previously found microglia to be overproduced, and was accompanied by an increase in presynaptic release probability and alterations in postsynaptic kinetics. Tissue-level NAc proteomics confirmed that microglial absence impacted numerous proteins involved in synapse structure, trans-synaptic signaling, and pre-synaptic function. However, microglial absence did not perturb levels of astrocyte-derived cues and adhesive proteins that promote synaptogenesis, suggesting that reduced synapse number may be caused by absence of a microglial-derived synaptogenic cue. Although observed electrophysiological synaptic changes were largely normalized by adulthood, we identified lasting effects of microglial absence on threat avoidance behavior, and these behavioral effects were directly associated with alterations of NAc neuronal activity. Together, these results indicate a critical role for microglia in regulating the synaptic landscape of the developing NAc and in establishing functional circuits underlying adult behavioral repertoires.
Collapse
Affiliation(s)
- Michael W Gongwer
- Department of Physiology, University of California Los Angeles, CA, USA
- Neuroscience Interdepartmental Program, University of California Los Angeles, CA, USA
- UCLA Medical Scientist Training Program, University of California Los Angeles, CA, USA
| | - Fanny Etienne
- Department of Physiology, University of California Los Angeles, CA, USA
| | - Eric N Moca
- Department of Physiology, University of California Los Angeles, CA, USA
| | - Megan S Chappell
- Department of Physiology, University of California Los Angeles, CA, USA
- Neuroscience Interdepartmental Program, University of California Los Angeles, CA, USA
| | - Sara V Blagburn-Blanco
- Department of Physiology, University of California Los Angeles, CA, USA
- Neuroscience Interdepartmental Program, University of California Los Angeles, CA, USA
- UCLA Medical Scientist Training Program, University of California Los Angeles, CA, USA
| | - Jack P Riley
- Department of Physiology, University of California Los Angeles, CA, USA
| | - Alexander S Enos
- Department of Physiology, University of California Los Angeles, CA, USA
| | - Melody Haratian
- Department of Physiology, University of California Los Angeles, CA, USA
| | - Alex Qi
- Department of Psychiatry, University of California Los Angeles, CA, USA
| | - Rocio Rojo
- Institution for Regeneration and Repair, University of Edinburgh, Scotland
| | - Scott A Wilke
- Department of Psychiatry, University of California Los Angeles, CA, USA
| | - Clare Pridans
- Institution for Regeneration and Repair, University of Edinburgh, Scotland
| | - Laura A DeNardo
- Department of Physiology, University of California Los Angeles, CA, USA
| | | |
Collapse
|
56
|
Jay TR, Kang Y, Ouellet-Massicotte V, Micael MKB, Kacouros-Perkins VL, Chen J, Sheehan A, Freeman MR. Developmental and age-related synapse elimination is mediated by glial Croquemort. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.24.600214. [PMID: 39026803 PMCID: PMC11257470 DOI: 10.1101/2024.06.24.600214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Neurons and glia work together to dynamically regulate neural circuit assembly and maintenance. In this study, we show Drosophila exhibit large-scale synapse formation and elimination as part of normal CNS circuit maturation, and that glia use conserved molecules to regulate these processes. Using a high throughput ELISA-based in vivo screening assay, we identify new glial genes that regulate synapse numbers in Drosophila in vivo, including the scavenger receptor ortholog Croquemort (Crq). Crq acts as an essential regulator of glial-dependent synapse elimination during development, with glial Crq loss leading to excess CNS synapses and progressive seizure susceptibility in adults. Loss of Crq in glia also prevents age-related synaptic loss in the adult brain. This work provides new insights into the cellular and molecular mechanisms that underlie synapse development and maintenance across the lifespan, and identifies glial Crq as a key regulator of these processes.
Collapse
|
57
|
Zhao Q, Li S, Krall L, Li Q, Sun R, Yin Y, Fu J, Zhang X, Wang Y, Yang M. Deciphering cellular complexity: advances and future directions in single-cell protein analysis. Front Bioeng Biotechnol 2025; 12:1507460. [PMID: 39877263 PMCID: PMC11772399 DOI: 10.3389/fbioe.2024.1507460] [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: 10/07/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025] Open
Abstract
Single-cell protein analysis has emerged as a powerful tool for understanding cellular heterogeneity and deciphering the complex mechanisms governing cellular function and fate. This review provides a comprehensive examination of the latest methodologies, including sophisticated cell isolation techniques (Fluorescence-Activated Cell Sorting (FACS), Magnetic-Activated Cell Sorting (MACS), Laser Capture Microdissection (LCM), manual cell picking, and microfluidics) and advanced approaches for protein profiling and protein-protein interaction analysis. The unique strengths, limitations, and opportunities of each method are discussed, along with their contributions to unraveling gene regulatory networks, cellular states, and disease mechanisms. The importance of data analysis and computational methods in extracting meaningful biological insights from the complex data generated by these technologies is also highlighted. By discussing recent progress, technological innovations, and potential future directions, this review emphasizes the critical role of single-cell protein analysis in advancing life science research and its promising applications in precision medicine, biomarker discovery, and targeted therapeutics. Deciphering cellular complexity at the single-cell level holds immense potential for transforming our understanding of biological processes and ultimately improving human health.
Collapse
Affiliation(s)
- Qirui Zhao
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Yunnan University, Kunming, China
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, China
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Shan Li
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Yunnan University, Kunming, China
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, China
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Leonard Krall
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Yunnan University, Kunming, China
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, China
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Qianyu Li
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Rongyuan Sun
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Yuqi Yin
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Jingyi Fu
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Xu Zhang
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Yunnan University, Kunming, China
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, China
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Yonghua Wang
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Yunnan University, Kunming, China
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, China
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Mei Yang
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Yunnan University, Kunming, China
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, China
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| |
Collapse
|
58
|
Haorah J, Malaroviyam S, Iyappan H, Samikkannu T. Neurological impact of HIV/AIDS and substance use alters brain function and structure. Front Med (Lausanne) 2025; 11:1505440. [PMID: 39839621 PMCID: PMC11747747 DOI: 10.3389/fmed.2024.1505440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025] Open
Abstract
Human immunodeficiency virus (HIV) infection is the cause of acquired immunodeficiency syndrome (AIDS). Combination antiretroviral therapy (cART) has successfully controlled AIDS, but HIV-associated neurocognitive disorders (HANDs) remain prevalent among people with HIV. HIV infection is often associated with substance use, which promotes HIV transmission and viral replication and exacerbates HANDs even in the era of cART. Thus, the comorbid effects of substance use exacerbate the neuropathogenesis of HANDs. Unraveling the mechanism(s) of this comorbid exacerbation at the molecular, cell-type, and brain region levels may provide a better understanding of HAND persistence. This review aims to highlight the comorbid effects of HIV and substance use in specific brain regions and cell types involved in the persistence of HANDs. This review includes an overview of post-translational modifications, alterations in microglia-specific biomarkers, and possible mechanistic pathways that may link epigenomic modifications to functional protein alterations in microglia. The impairment of the microglial proteins that are involved in neural circuit function appears to contribute to the breakdown of cellular communication and neurodegeneration in HANDs. The epigenetic modification of N-terminal acetylation is currently understudied, which is discussed in brief to demonstrate the important role of this epigenetic modification in infected microglia within specific brain regions. The discussion also explores whether combined antiretroviral therapy is effective in preventing HIV infection or substance-use-mediated post-translational modifications and protein alterations in the persistence of neuropathogenesis in HANDs.
Collapse
Affiliation(s)
| | | | | | - Thangavel Samikkannu
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, College Station, TX, United States
| |
Collapse
|
59
|
Vella VR, Holman PJ, Bodnar TS, Raineki C. Ontogenetic Neuroimmune Changes Following Prenatal Alcohol Exposure: Implications for Neurobehavioral Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1473:15-39. [PMID: 40128473 DOI: 10.1007/978-3-031-81908-7_2] [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: 03/26/2025]
Abstract
This chapter reviews the enduring effects of prenatal alcohol exposure (PAE) on neuroimmune function across the lifespan, including discussion of associated neurobehavioral alterations. Alcohol has potent teratogenic effects, with a large body of work linking PAE to perturbations in neuroimmune function. These PAE-related neuroimmune disturbances may have downstream effects on neurobehavioral function given the critical role of the neuroimmune system in central nervous system development. The neuroimmune system matures over time, playing distinct roles depending on the developmental processes occurring within that maturational stage. This chapter thus takes an ontogenetic approach to understanding how PAE induces unique neuroimmune changes across the lifespan, beginning with a review of changes in early life before moving into adolescence and ending in adulthood. The focus will be on work utilizing rodent models, which allow for more tightly controlled conditions than are possible in human research. The chapter concludes with a discussion of possible mechanisms underlying the developmental changes in neuroimmune function following PAE, with a specific focus on the role of the gut microbiota.
Collapse
Affiliation(s)
- Victoria R Vella
- Department of Psychology, Brock University, St. Catharines, ON, Canada
| | - Parker J Holman
- Department of Psychology, Brock University, St. Catharines, ON, Canada
| | - Tamara S Bodnar
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, Calgary, AB, Canada
- Hotchkiss Brain Institute, Calgary, AB, Canada
| | - Charlis Raineki
- Department of Psychology, Brock University, St. Catharines, ON, Canada.
- Centre for Neuroscience, Brock University, St. Catharines, ON, Canada.
| |
Collapse
|
60
|
Chen CH, Hsu SY, Yu WJ, Chiang CS, Yu CF. Distinct roles of small extracellular vesicles from resident and infiltrating macrophages on glioma growth and mobility. J Cancer 2025; 16:969-981. [PMID: 39781357 PMCID: PMC11705045 DOI: 10.7150/jca.103595] [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: 09/12/2024] [Accepted: 12/15/2024] [Indexed: 01/12/2025] Open
Abstract
Previous studies revealed that tumor-associated macrophages/microglia (TAMs) promoted glioma invasiveness during tumor progression and after radiotherapy. However, the communication of TAMs with tumor cells remains unclear. This study aimed to examine the role of small extracellular vesicles (sEVs) derived from TAMs in TAMs-mediated brain tumor invasion. This study utilized BV2 and RAW264.7 cell lines representing resident and infiltrating macrophages, respectively, to unveil their effect on tumor cells. Purified sEVs from BV2 and RAW264.7 were validated by nanoparticle track analysis (NTA), transmission electron microscopy (TEM), and western blotting for sEV markers. The effect of sEVs on the murine astrocytoma tumor cell line ALTS1C1 was examined on cell proliferation, migration, and gene expression. The results showed that ALTS1C1 cells effectively engulfed sEVs purified from BV2 and RAW264.7. Only BV2-derived sEVs promoted cell proliferation and were dose-dependent. Further, morphological changes in ALTS1C1 cells were observed after incubation with BV2-derived sEVs, which was associated with enhancing cell migration. BV2-mediated glioma proliferation and mobility were related to the upregulation of vascular endothelial growth factor (VEGF) and downregulation of death effector domain-containing protein (DEDD) gene expression. This study demonstrates the distinct function of sEVs of resident macrophages on glioma cell invasion and reveals the mechanism underlying microglia-mediated tumor progression. These findings suggested resident microglia is the potential therapeutic target for TAMs-induced brain tumor invasiveness.
Collapse
Affiliation(s)
- Chu-Hsuan Chen
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsin-Chu, Taiwan
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsin-Chu, Taiwan
| | - Sheng-Yun Hsu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsin-Chu, Taiwan
| | - Wen-Jui Yu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsin-Chu, Taiwan
| | - Chi-Shiun Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsin-Chu, Taiwan
- Institute of Nuclear Engineering and Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
- The BNCT Research Center, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Ching-Fang Yu
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, 33302, Taiwan
- Research Center for Radiation Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Radiation Oncology, Chang Gung Memorial Hospital Linkou Branch, Taoyuan 33382, Taiwan
| |
Collapse
|
61
|
Chauveau F, Winkeler A, Chalon S, Boutin H, Becker G. PET imaging of neuroinflammation: any credible alternatives to TSPO yet? Mol Psychiatry 2025; 30:213-228. [PMID: 38997465 DOI: 10.1038/s41380-024-02656-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024]
Abstract
Over the last decades, the role of neuroinflammation in neuropsychiatric conditions has attracted an exponentially growing interest. A key driver for this trend was the ability to image brain inflammation in vivo using PET radioligands targeting the Translocator Protein 18 kDa (TSPO), which is known to be expressed in activated microglia and astrocytes upon inflammatory events as well as constitutively in endothelial cells. TSPO is a mitochondrial protein that is expressed mostly by microglial cells upon activation but is also expressed by astrocytes in some conditions and constitutively by endothelial cells. Therefore, our current understanding of neuroinflammation dynamics is hampered by the lack of alternative targets available for PET imaging. We performed a systematic search and review on radiotracers developed for neuroinflammation PET imaging apart from TSPO. The following targets of interest were identified through literature screening (including previous narrative reviews): P2Y12R, P2X7R, CSF1R, COX (microglial targets), MAO-B, I2BS (astrocytic targets), CB2R & S1PRs (not specific of a single cell type). We determined the level of development and provided a scoping review for each target. Strikingly, astrocytic biomarker MAO-B has progressed in clinical investigations the furthest, while few radiotracers (notably targeting S1P1Rs, CSF1R) are being implemented in clinical investigations. Other targets such as CB2R and P2X7R have proven disappointing in clinical studies (e.g. poor signal, lack of changes in disease conditions, etc.). While astrocytic targets are promising, development of new biomarkers and tracers specific for microglial activation has proven challenging.
Collapse
Affiliation(s)
- Fabien Chauveau
- Université Claude Bernard Lyon 1, Centre de Recherche en Neurosciences de Lyon, Inserm U1028, CNRS UMR5292, BIORAN, Groupement Hospitalier Est - CERMEP, 59 boulevard Pinel, 69677, Bron, Cedex, France
| | - Alexandra Winkeler
- Université Paris-Saclay, Inserm, CNRS, CEA, BioMaps, Service Hospitalier Frédéric Joliot, 4 place du général Leclerc, 91401, Orsay, France
| | - Sylvie Chalon
- UMR 1253 iBrain, Université de Tours - INSERM, Bâtiment Planiol, UFR de Médecine, 10 Boulevard Tonnellé, 37032, Tours, Cedex 01, France
| | - Hervé Boutin
- UMR 1253 iBrain, Université de Tours - INSERM, Bâtiment Planiol, UFR de Médecine, 10 Boulevard Tonnellé, 37032, Tours, Cedex 01, France.
| | - Guillaume Becker
- Université Claude Bernard Lyon 1, Centre de Recherche en Neurosciences de Lyon, Inserm U1028, CNRS UMR5292, BIORAN, Groupement Hospitalier Est - CERMEP, 59 boulevard Pinel, 69677, Bron, Cedex, France
- Agence Nationale de Sécurité Sanitaire de l'Alimentation, de l'Environnement et du Travail, 14 rue Pierre et Marie Curie, 94701, Maisons-Alfort, Cedex, France
| |
Collapse
|
62
|
Gence L, Morello E, Rastegar S, Apalama ML, Meilhac O, Bascands JL, Diotel N. Gene expression patterns of the LDL receptor and its inhibitor Pcsk9 in the adult zebrafish brain suggest a possible role in neurogenesis. Eur J Neurosci 2025; 61:e16586. [PMID: 39551948 DOI: 10.1111/ejn.16586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 09/18/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024]
Abstract
The low-density lipoprotein receptor (LDLr) is the first member of a closely related transmembrane protein family. It is known for its involvement in various physiological processes, mainly in the regulation of lipid metabolism, especially in the brains of mammals and zebrafish. In zebrafish, two ldlr genes (ldlra and b) have been identified and their distribution in the brain is not well documented. Recently, the roles of ldlr and its inhibitor pcsk9 in regenerative process after telencephalic brain injury have been discussed. In this study, we explored the expression patterns of these genes during zebrafish development. We found that ldlra expression was detected at the end of the pharyngula period (48 hpf) and increased during the larval stage. Conversely, ldlrb expression was observed from zygotic to larval stages. Using techniques like in situ hybridization and taking advantage of transgenic fish, we demonstrated the widespread distribution of ldlra, ldlrb and pcsk9 in the brain of adult zebrafish. Specifically, these genes were expressed in neurons and neural stem cells and also at lower levels in endothelial cells. As expected, intraperitoneal injection of fluorescent-labelled LDLs resulted in their uptake by cerebral endothelial cells in a homeostatic context, whereas they diffused within the brain parenchyma after telencephalic injury. However, after intracerebroventricular injections into animals, LDL particles were not taken up by neural stem cells. In conclusion, our results provide additional evidence for LDLr expression in the brain of adult zebrafish. These results raise the question of the role of LDLr in the cholesterol/lipid imbalance in cerebral complications.
Collapse
Affiliation(s)
- Laura Gence
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Pierre, La Réunion, France
| | - Elena Morello
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Pierre, La Réunion, France
- CHU de La Réunion, Saint-Pierre, La Réunion, France
| | - Sepand Rastegar
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Marie Laurine Apalama
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Pierre, La Réunion, France
| | - Olivier Meilhac
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Pierre, La Réunion, France
- CHU de La Réunion, Saint-Pierre, La Réunion, France
| | - Jean-Loup Bascands
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Pierre, La Réunion, France
| | - Nicolas Diotel
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Pierre, La Réunion, France
| |
Collapse
|
63
|
Traetta ME, Vecchiarelli HA, Tremblay MÈ. Fundamental Neurochemistry Review: Lipids across microglial states. J Neurochem 2025; 169:e16259. [PMID: 39696753 DOI: 10.1111/jnc.16259] [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: 08/08/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 12/20/2024]
Abstract
The capacity of immune cells to alter their function based on their metabolism is the basis of the emerging field of immunometabolism. Microglia are the resident innate immune cells of the central nervous system, and it is a current focus of the field to investigate how alterations in their metabolism impact these cells. Microglia have the ability to utilize lipids, such as fatty acids, as energy sources, but also alterations in lipids can impact microglial form and function. Recent studies highlighting different microglial states and transcriptional signatures have highlighted modifications in lipid processing as defining these states. This review highlights these recent studies and uses these altered pathways to discuss the current understanding of lipid biology in microglia. The studies highlighted here review how lipids may alter microglial phagocytic functioning or alter their pro- and anti-inflammatory balance. These studies provide a foundation by which lipid supplementation or diet alterations could influence microglial states and function. Furthermore, targets modulating microglial lipid metabolism may provide new treatment avenues.
Collapse
Affiliation(s)
- Marianela E Traetta
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Haley A Vecchiarelli
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, British Columbia, Canada
- Institute for Aging and Lifelong Health (IALH), University of Victoria, Victoria, British Columbia, Canada
- Département de médecine moléculaire, Université Laval, Québec City, Quebec, Canada
- Axe neurosciences, Centre de recherche du CHU de Québec, Université Laval, Québec City, Quebec, Canada
- Neurology and Neurosurgery Department, McGill University, Montréal, Quebec, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
64
|
Gobbo D, Kirchhoff F. Animal-based approaches to understanding neuroglia physiology in vitro and in vivo. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:229-263. [PMID: 40122627 DOI: 10.1016/b978-0-443-19104-6.00012-7] [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: 03/25/2025]
Abstract
This chapter describes the pivotal role of animal models for unraveling the physiology of neuroglial cells in the central nervous system (CNS). The two rodent species Mus musculus (mice) and Rattus norvegicus (rats) have been indispensable in scientific research due to their remarkable resemblance to humans anatomically, physiologically, and genetically. Their ease of maintenance, short gestation times, and rapid development make them ideal candidates for studying the physiology of astrocytes, oligodendrocyte-lineage cells, and microglia. Moreover, their genetic similarity to humans facilitates the investigation of molecular mechanisms governing neural physiology. Mice are largely the predominant model of neuroglial research, owing to advanced genetic manipulation techniques, whereas rats remain invaluable for applications requiring larger CNS structures for surgical manipulations. Next to rodents, other animal models, namely, Danio rerio (zebrafish) and Drosophila melanogaster (fruit fly), will be discussed to emphasize their critical role in advancing our understanding of glial physiology. Each animal model provides distinct advantages and disadvantages. By combining the strengths of each of them, researchers can gain comprehensive insights into glial function across species, ultimately promoting the understanding of glial physiology in the human CNS and driving the development of novel therapeutic interventions for CNS disorders.
Collapse
Affiliation(s)
- Davide Gobbo
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany.
| | - Frank Kirchhoff
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany; Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, Homburg, Germany.
| |
Collapse
|
65
|
Latham AS, Geer CE, Ackart DF, Weninger KN, Gross CC, Podell BK, Basaraba RJ, Moreno JA. Immune cell infiltration and modulation of the blood-brain barrier in a guinea pig model of tuberculosis: Observations without evidence of bacterial dissemination to the brain. PLoS One 2024; 19:e0307577. [PMID: 39739680 DOI: 10.1371/journal.pone.0307577] [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: 09/05/2023] [Accepted: 07/08/2024] [Indexed: 01/02/2025] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) infection, is a chronic inflammatory disease. Although typically associated with inflammation of the lungs and other peripheral tissues, increasing evidence has uncovered neurological consequences attributable to Mtb infection. These include deficits in memory and cognition, increased risk for neurodegenerative disease, and progressive neuropathology. Although the neurological effects of the disease, without CNS infection, have been characterized, the mechanism of neurotoxicity is unknown. We hypothesized that alterations to the blood-brain barrier (BBB) allows peripheral immune cells to enter the brain, initiating a neuroinflammatory response. To test this hypothesis, guinea pigs were exposed by aerosol to a laboratory and a clinical Mtb strain for 15 days. Following Mtb infection, proteins critical to BBB function, including claudin V and collagen IV, are modulated without evidence of bacterial dissemination to the brain. This is correlated with increased contact of astrocytic processes to vessels in the brain, as well as increased expression of the water channel protein aquaporin 4 (AQP4) on endfeet. Upon further investigation, we discovered the potential role of glial reactivity, which is increased following infection with both bacterial strains, in the progression of BBB changes and, ultimately, the permeability of peripheral immune cells into the brain. Through these data, we have obtained a preliminary understanding of the mechanisms of cellular stress in the brain following pulmonary Mtb infection which should be further investigated in future studies.
Collapse
Affiliation(s)
- Amanda S Latham
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
- Brain Research Center, Colorado State University, Fort Collins, Colorado, United States of America
| | - Charlize E Geer
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - David F Ackart
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Kristin N Weninger
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
- Department of Biomedical Science, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Chase C Gross
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Brendan K Podell
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Randall J Basaraba
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Julie A Moreno
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
- Brain Research Center, Colorado State University, Fort Collins, Colorado, United States of America
- Center for Healthy Aging, Colorado State University, Fort Collins, Colorado, United States of America
| |
Collapse
|
66
|
Lv X, Jia M, Feng X, Jian JX, Yang JJ, Ma DQ, Ji MH, Diao YG, Shen JC. STING Driving Synaptic Phagocytosis of Hippocampal Microglia/Macrophages Contributes to Cognitive Impairment in Sepsis-Associated Encephalopathy in Mice. CNS Neurosci Ther 2024; 30:e70166. [PMID: 39699038 DOI: 10.1111/cns.70166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 09/21/2024] [Accepted: 11/23/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Sepsis-associated encephalopathy (SAE) is a serious neurologic complication in septic patients with poor prognoses. There is increasing evidence that stimulator of interferon genes (STING) plays a crucial role in neuroinflammation and cognitive impairment. However, whether sepsis associated with STING changes contributes to cognitive impairment is unknown. METHODS Male adult mice received lipopolysaccharide (LPS) injection (a single dose of 4 mg/kg; i.p. injection) and 30 min later, they were injected with STING inhibitor C-176 (a single dose of 30 mg/kg, i.p. injection). Behavioral assessments, biochemical measurements, in vivo and ex vivo electrophysiology techniques were conducted to investigate the association between LPS-induced STING overexpression and cognitive function. RESULTS Cognitive impairment was associated with STING overexpression and activation of microglia/macrophages. Phagocytosis of microglia/macrophages as well as complement C1q release were increased after LPS injection, leading to abnormal pruning synapses, synaptic transmission reduction, long-term potentiation (LTP) impairment, as well as abnormal theta oscillation in the hippocampus. Notably, STING inhibitor C-176 significantly reversed these changes. CONCLUSIONS Sepsis-induced STING overexpression in microglia/macrophages may lead to synaptic loss, abnormal theta oscillation and LTP impairment through microglia/macrophages activation and complement C1q modulation, ultimately resulting in cognitive impairment.
Collapse
Affiliation(s)
- Xin Lv
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Anesthesiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Min Jia
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiao Feng
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jia-Xiong Jian
- Department of Anesthesiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jian-Jun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Da-Qing Ma
- Division of Anaesthetics, Pain Medicine & Intensive Care, Department of Surgery & Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
- Perioperative and Systems Medicine Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Mu-Huo Ji
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu-Gang Diao
- Department of Anesthesiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Jin-Chun Shen
- Department of Anesthesiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
67
|
Sabogal-Guaqueta AM, Mitchell-Garcia T, Hunneman J, Voshart D, Thiruvalluvan A, Foijer F, Kruyt F, Trombetta-Lima M, Eggen BJL, Boddeke E, Barazzuol L, Dolga AM. Brain organoid models for studying the function of iPSC-derived microglia in neurodegeneration and brain tumours. Neurobiol Dis 2024; 203:106742. [PMID: 39581553 DOI: 10.1016/j.nbd.2024.106742] [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: 09/30/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024] Open
Abstract
Microglia represent the main resident immune cells of the brain. The interplay between microglia and other cells in the central nervous system, such as neurons or other glial cells, influences the function and ability of microglia to respond to various stimuli. These cellular communications, when disrupted, can affect the structure and function of the brain, and the initiation and progression of neurodegenerative diseases including Alzheimer's disease and Parkinson's disease, as well as the progression of other brain diseases like glioblastoma. Due to the difficult access to patient brain tissue and the differences reported in the murine models, the available models to study the role of microglia in disease progression are limited. Pluripotent stem cell technology has facilitated the generation of highly complex models, allowing the study of control and patient-derived microglia in vitro. Moreover, the ability to generate brain organoids that can mimic the 3D tissue environment and intercellular interactions in the brain provide powerful tools to study cellular pathways under homeostatic conditions and various disease pathologies. In this review, we summarise the most recent developments in modelling degenerative diseases and glioblastoma, with a focus on brain organoids with integrated microglia. We provide an overview of the most relevant research on intercellular interactions of microglia to evaluate their potential to study brain pathologies.
Collapse
Affiliation(s)
- Angelica Maria Sabogal-Guaqueta
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands.
| | - Teresa Mitchell-Garcia
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands
| | - Jasmijn Hunneman
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands
| | - Daniëlle Voshart
- Department of Biomedical Sciences, Section of Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands; Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Arun Thiruvalluvan
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Floris Foijer
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Frank Kruyt
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marina Trombetta-Lima
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands; Faculty of Science and Engineering, Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands
| | - Bart J L Eggen
- Department of Biomedical Sciences, Section of Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| | - Erik Boddeke
- Department of Biomedical Sciences, Section of Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| | - Lara Barazzuol
- Department of Biomedical Sciences, Section of Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands; Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Amalia M Dolga
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands; Department Pathology and Medical biology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
68
|
Solomou G, Young AMH, Bulstrode HJCJ. Microglia and macrophages in glioblastoma: landscapes and treatment directions. Mol Oncol 2024; 18:2906-2926. [PMID: 38712663 PMCID: PMC11619806 DOI: 10.1002/1878-0261.13657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/29/2024] [Accepted: 04/19/2024] [Indexed: 05/08/2024] Open
Abstract
Glioblastoma is the most common primary malignant tumour of the central nervous system and remains uniformly and rapidly fatal. The tumour-associated macrophage (TAM) compartment comprises brain-resident microglia and bone marrow-derived macrophages (BMDMs) recruited from the periphery. Immune-suppressive and tumour-supportive TAM cell states predominate in glioblastoma, and immunotherapies, which have achieved striking success in other solid tumours have consistently failed to improve survival in this 'immune-cold' niche context. Hypoxic and necrotic regions in the tumour core are found to enrich, especially in anti-inflammatory and immune-suppressive TAM cell states. Microglia predominate at the invasive tumour margin and express pro-inflammatory and interferon TAM cell signatures. Depletion of TAMs, or repolarisation towards a pro-inflammatory state, are appealing therapeutic strategies and will depend on effective understanding and classification of TAM cell ontogeny and state based on new single-cell and spatial multi-omic in situ profiling. Here, we explore the application of these datasets to expand and refine TAM characterisation, to inform improved modelling approaches, and ultimately underpin the effective manipulation of function.
Collapse
Affiliation(s)
- Georgios Solomou
- Wellcome MRC Cambridge Stem Cell InstituteUniversity of CambridgeUK
- Department of NeurosurgeryAddenbrooke's HospitalCambridgeUK
| | - Adam M. H. Young
- Wellcome MRC Cambridge Stem Cell InstituteUniversity of CambridgeUK
- Department of NeurosurgeryAddenbrooke's HospitalCambridgeUK
| | - Harry J. C. J. Bulstrode
- Wellcome MRC Cambridge Stem Cell InstituteUniversity of CambridgeUK
- Department of NeurosurgeryAddenbrooke's HospitalCambridgeUK
| |
Collapse
|
69
|
Bastiancich C, Snacel-Fazy E, Fernandez S, Robert S, Stacchini R, Plantureux L, Boissonneau S, Testud B, Guillet B, Debarbieux F, Luche H, Figarella-Branger D, Estève MA, Tabouret E, Tchoghandjian A. Tailoring glioblastoma treatment based on longitudinal analysis of post-surgical tumor microenvironment. J Exp Clin Cancer Res 2024; 43:311. [PMID: 39605004 PMCID: PMC11603899 DOI: 10.1186/s13046-024-03231-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024] Open
Abstract
Glioblastoma (GBM), an incurable primary brain tumor, typically requires surgical intervention followed by chemoradiation; however, recurrences remain fatal. Our previous work demonstrated that a nanomedicine hydrogel (GemC12-LNC) delays recurrence when administered post-surgery. However, tumor debulking also triggers time-dependent immune reactions that promote recurrence at the resection cavity borders. We hypothesized that combining the hydrogel with an immunomodulatory drug could enhance therapeutic outcomes. A thorough characterization of the post-surgical microenvironment (SMe) is crucial to guide combinatorial approaches.In this study, we performed cellular resolution imaging, flow cytometry and spatial hyperplexed immunofluorescence imaging to characterize the SMe in a syngeneic mouse model of tumor resection. Owing to our dynamic approach, we observed transient opening of the blood-brain barrier (BBB) during the first week after surgery. BBB permeability post-surgery was also confirmed in GBM patients. In our murine model, we also observed changes in immune cell morphology and spatial location post-surgery over time in resected animals as well as the accumulation of reactive microglia and anti-inflammatory macrophages in recurrences compared to unresected tumors since the first steps of recurrence growth. Therefore we investigated whether starting a systemic treatment with the SMAC mimetic small molecule (GDC-0152) directly after surgery would be beneficial for enhancing microglial anti-tumoral activity and decreasing the number of anti-inflammatory macrophages around the GemC12-LNC hydrogel-loaded tumor cavity. The immunomodulatory effects of this drug combination was firstly shown in patient-derived tumoroids. Its efficacy was confirmed in vivo by survival analysis and correlated with reversal of the immune profile as well as delayed tumor recurrence.This comprehensive study identified critical time frames and immune cellular targets within the SMe, aiding in the rational design of combination therapies to delay recurrence onset. Our findings suggest that post-surgical systemic injection of GDC-0152 in combination with GemC12-LNC local treatment is a promising and innovative approach for managing GBM recurrence, with potential for future translation to human patient.
Collapse
Affiliation(s)
- Chiara Bastiancich
- Aix-Marseille Univ, CNRS, INP, Institute of Neurophysiopathology UMR7051, Team Gliomagenesis and Microenvironment, Faculté des Sciences Médicales et Paramédicales - Secteur Timone, 27, Bd Jean Moulin, Marseille, 13005, France.
- Department of Drug Science and Technology, University of Turin, Turin, 10125, Italy.
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73, Brussels, 1200, Belgium.
- Aix-Marseille Univ, Réseau Préclinique Et Translationnel de Recherche en Neuro-Oncologie, Plateforme PETRA"TECH", Marseille, 13005, France.
| | - Emmanuel Snacel-Fazy
- Aix-Marseille Univ, CNRS, INP, Institute of Neurophysiopathology UMR7051, Team Gliomagenesis and Microenvironment, Faculté des Sciences Médicales et Paramédicales - Secteur Timone, 27, Bd Jean Moulin, Marseille, 13005, France
| | | | | | - Roberta Stacchini
- Aix-Marseille Univ, CNRS, INP, Institute of Neurophysiopathology UMR7051, Team Gliomagenesis and Microenvironment, Faculté des Sciences Médicales et Paramédicales - Secteur Timone, 27, Bd Jean Moulin, Marseille, 13005, France
| | - Léa Plantureux
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
| | - Sébastien Boissonneau
- Department of Neuro-Surgery, AP-HM, Hôpital Universitaire Timone, Marseille, 13005, France
- Department of Neuro-Surgery, Valenciennes Hospital, Valenciennes, 59300, France
| | - Benoit Testud
- Aix Marseille Univ, CNRS, CRMBM, Marseille, France
- Aix Marseille Univ, APHM, Hôpital Universitaire Timone, CEMEREM, Marseille, 13005, France
- Department of Neuroradiology, Aix Marseille Univ, APHM, Hôpital Universitaire Timone, Marseille, 13005, France
| | - Benjamin Guillet
- Aix Marseille Univ, CNRS, CERIMED, Marseille, France
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
- Aix Marseille Univ, APHM, Hôpital Timone, Pôle Pharmacie, Radiopharmacie, Marseille, 13005, France
| | - Franck Debarbieux
- Aix Marseille Univ, CNRS, CERIMED, Marseille, France
- Aix Marseille Univ, CNRS, INT, Inst Neurosci Timone, Marseille, France
- Institut Universitaire de France, Paris, 75005, France
| | - Hervé Luche
- Aix-Marseille Univ, CNRS, INSERM, CIPHE, Marseille, 13009, France
| | - Dominique Figarella-Branger
- Aix-Marseille Univ, CNRS, INP, Institute of Neurophysiopathology UMR7051, Team Gliomagenesis and Microenvironment, Faculté des Sciences Médicales et Paramédicales - Secteur Timone, 27, Bd Jean Moulin, Marseille, 13005, France
| | - Marie-Anne Estève
- Aix-Marseille Univ, CNRS, INP, Institute of Neurophysiopathology UMR7051, Team Gliomagenesis and Microenvironment, Faculté des Sciences Médicales et Paramédicales - Secteur Timone, 27, Bd Jean Moulin, Marseille, 13005, France
- Aix Marseille Univ, APHM, Hôpital Timone, Service Pharmacie, Marseille, 13005, France
| | - Emeline Tabouret
- Aix-Marseille Univ, CNRS, INP, Institute of Neurophysiopathology UMR7051, Team Gliomagenesis and Microenvironment, Faculté des Sciences Médicales et Paramédicales - Secteur Timone, 27, Bd Jean Moulin, Marseille, 13005, France
- AP-HM, CHU Timone, Service de Neurooncologie, Marseille, France
- Aix-Marseille Univ, Réseau Préclinique Et Translationnel de Recherche en Neuro-Oncologie, Plateforme PE"TRANSLA", Marseille, 13005, France
| | - Aurélie Tchoghandjian
- Aix-Marseille Univ, CNRS, INP, Institute of Neurophysiopathology UMR7051, Team Gliomagenesis and Microenvironment, Faculté des Sciences Médicales et Paramédicales - Secteur Timone, 27, Bd Jean Moulin, Marseille, 13005, France.
- Aix-Marseille Univ, Réseau Préclinique Et Translationnel de Recherche en Neuro-Oncologie, Plateforme PETRA"TECH", Marseille, 13005, France.
| |
Collapse
|
70
|
Bhatt M, Sharma M, Das B. The Role of Inflammatory Cascade and Reactive Astrogliosis in Glial Scar Formation Post-spinal Cord Injury. Cell Mol Neurobiol 2024; 44:78. [PMID: 39579235 PMCID: PMC11585509 DOI: 10.1007/s10571-024-01519-9] [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: 08/12/2024] [Accepted: 11/15/2024] [Indexed: 11/25/2024]
Abstract
Reactive astrogliosis and inflammation are pathologic hallmarks of spinal cord injury. After injury, dysfunction of glial cells (astrocytes) results in glial scar formation, which limits neuronal regeneration. The blood-spinal cord barrier maintains the structural and functional integrity of the spinal cord and does not allow blood vessel components to leak into the spinal cord microenvironment. After the injury, disruption in the spinal cord barrier causes an imbalance of the immunological microenvironment. This triggers the process of neuroinflammation, facilitated by the actions of microglia, neutrophils, glial cells, and cytokines production. Recent work has revealed two phenotypes of astrocytes, A1 and A2, where A2 has a protective type, and A1 releases neurotoxins, further promoting glial scar formation. Here, we first describe the current understanding of the spinal cord microenvironment, both pre-, and post-injury, and the role of different glial cells in the context of spinal cord injury, which forms the essential update on the cellular and molecular events following injury. We aim to explore in-depth signaling pathways and molecular mediators that trigger astrocyte activation and glial scar formation. This review will discuss the activated signaling pathways in astrocytes and other glial cells and their collaborative role in the development of gliosis through inflammatory responses.
Collapse
Affiliation(s)
- Manini Bhatt
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Bara Phool, Punjab, India
| | - Muskan Sharma
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Bara Phool, Punjab, India
| | - Bodhisatwa Das
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Bara Phool, Punjab, India.
| |
Collapse
|
71
|
Althammer F, Roy RK, Kirchner MK, Podpecan Y, Helen J, McGrath S, Lira EC, Stern JE. Angiotensin-II drives changes in microglia-vascular interactions in rats with heart failure. Commun Biol 2024; 7:1537. [PMID: 39562706 PMCID: PMC11577102 DOI: 10.1038/s42003-024-07229-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
Activation of microglia, the resident immune cells of the central nervous system, leading to the subsequent release of pro-inflammatory cytokines, has been linked to cardiac remodeling, autonomic disbalance, and cognitive deficits in heart failure (HF). While previous studies emphasized the role of hippocampal Angiotensin II (AngII) signaling in HF-induced microglial activation, unanswered mechanistic questions persist. Evidence suggests significant interactions between microglia and local microvasculature, potentially affecting blood-brain barrier integrity and cerebral blood flow regulation. Still, whether the microglial-vascular interface is affected in the brain during HF remains unknown. Using a well-established ischemic HF rat model, we demonstrate the increased abundance of vessel-associated microglia (VAM) in HF rat hippocampi, along with an increased expression of AngII AT1a receptors. Acute AngII administration to sham rats induced microglia recruitment to brain capillaries, along with increased expression of TNFα. Conversely, administering an AT1aR blocker to HF rats prevented the recruitment of microglia to blood vessels, normalizing their levels to those in healthy rats. These results highlight the critical importance of a rather understudied phenomenon (i.e., microglia-vascular interactions in the brain) in the context of the pathophysiology of a highly prevalent cardiovascular disease, and unveil novel potential therapeutic avenues aimed at mitigating neuroinflammation in cardiovascular diseases.
Collapse
Affiliation(s)
- Ferdinand Althammer
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
- Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| | - Ranjan K Roy
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Matthew K Kirchner
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Yuval Podpecan
- Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| | - Jemima Helen
- Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| | - Shaina McGrath
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Elba Campos Lira
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Javier E Stern
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA.
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
72
|
Li H, Hu W, Wu Z, Tian B, Ren Y, Zou X. Esketamine improves cognitive function in sepsis-associated encephalopathy by inhibiting microglia-mediated neuroinflammation. Eur J Pharmacol 2024; 983:177014. [PMID: 39312992 DOI: 10.1016/j.ejphar.2024.177014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 09/14/2024] [Accepted: 09/20/2024] [Indexed: 09/25/2024]
Abstract
Microglia-mediated neuroinflammation is critical in the pathogenesis of sepsis-associated encephalopathy(SAE). Identifying the key factors that inhibit microglia-mediated neuroinflammation holds promise as a potential target for preventing and treating SAE. Esketamine, a non-competitive N-methyl-D-aspartate (NMDA) receptor antagonist, has been proposed to possess protective and therapeutic properties against neuroinflammatory disorders. This study provides evidence that the administration of Esketamine in SAE mice improves cognitive impairments and alleviates neuronal damage by inhibiting the microglia-mediated neuroinflammation. The BDNF receptor antagonist K252a was employed in both vivo and in vitro experiments. The findings indicate that K252a successfully counteracted the beneficial effects of Esketamine on microglia and cognitive behavior in mice with SAE. Consequently, these results suggest that Esketamine inhibits microglia-mediated neuroinflammation by activating the BDNF pathway, and mitigating neuronal damage and cognitive dysfunction associated with SAE.
Collapse
Affiliation(s)
- Hui Li
- College of Anesthesia, Guizhou Medical University, Guizhou Province, Guiyang 550004, China; Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guizhou Province, Guiyang 550004, China; Guizhou Medical University Key Laboratory of Anesthesia and Pain Mechanism Research, Guizhou Province, Guiyang, 550004, China
| | - Wen Hu
- College of Anesthesia, Guizhou Medical University, Guizhou Province, Guiyang 550004, China; Guizhou Medical University Key Laboratory of Anesthesia and Pain Mechanism Research, Guizhou Province, Guiyang, 550004, China
| | - Zhen Wu
- College of Anesthesia, Guizhou Medical University, Guizhou Province, Guiyang 550004, China; Guizhou Medical University Key Laboratory of Anesthesia and Pain Mechanism Research, Guizhou Province, Guiyang, 550004, China
| | - Bin Tian
- Department of Radiology, The Second People's Hospital of Guiyang, Guizhou Province, Guiyang, 550023, China
| | - Yimin Ren
- College of Anesthesia, Guizhou Medical University, Guizhou Province, Guiyang 550004, China; Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guizhou Province, Guiyang 550004, China; Guizhou Medical University Key Laboratory of Anesthesia and Pain Mechanism Research, Guizhou Province, Guiyang, 550004, China.
| | - Xiaohua Zou
- College of Anesthesia, Guizhou Medical University, Guizhou Province, Guiyang 550004, China; Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guizhou Province, Guiyang 550004, China; Guizhou Medical University Key Laboratory of Anesthesia and Pain Mechanism Research, Guizhou Province, Guiyang, 550004, China.
| |
Collapse
|
73
|
Potthoff TE, Walter C, Jeising D, Münter D, Verma A, Suero Molina E, Stummer W, Dugas M, Hartmann W, Dottermusch M, Altendorf L, Schüller U, Scheuermann S, Seitz C, Albert TK, Kerl K. Single-cell transcriptomics link gene expression signatures to clinicopathological features of gonadotroph and lactotroph PitNET. J Transl Med 2024; 22:1027. [PMID: 39548496 PMCID: PMC11566263 DOI: 10.1186/s12967-024-05821-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 10/31/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Pituitary neuroendocrine tumors (PitNET) are among the most common intracranial tumors. Despite a frequent benign course, aggressive behavior can occur. Tumor behavior is known to be under the influence of the tumor microenvironment (TME). However, the relationship between TME cells and aggressive tumor behavior has not been adequately explored in PitNET. METHODS We performed differential expression analysis as well as gene expression program identification based on single-cell RNA sequencing to comparatively characterize the transcriptome of seven gonadotroph and three lactotroph PitNET and correlate it with clinical features using bulk RNA-seq data from an independent cohort of 134 PitNET. Tumor immune infiltration was quantified via immunostaining on tissue sections of gonadotroph and lactotroph PitNET. RESULTS In lactotroph PitNET, we detect a highly proliferative gene profile with significantly increased expression levels in aggressively growing tumors within bulk RNA-seq data of an independent cohort of 134 PitNET samples. We also report high intratumoral heterogeneity in gonadotroph PitNET (GoPN) and lactotroph PitNET (LaPN) and identify signatures of epithelial, endocrine, and immunological gene networks in both subtypes. A comparison of their TME composition shows enrichment of SPP1+ macrophages and CD4+ T cells in GoPN, as well as enrichment of CD4/CD8 double-negative T cells (DN) and natural killer cells (NK) in LaPN. Also notable is the presence of proliferative lymphocytes, the occurrence of which positively correlates with more aggressive tumor behavior in the bulk RNA-seq cohort. However, increased CD8+ T and NK cell abundances correlate significantly with reduced aggressiveness indicating potential anti-tumoral effects. CONCLUSIONS Our study expands the knowledge of the differences in cellular composition of gonadotroph and lactotroph PitNET subtypes. It lays the foundation for further studies on the influence of lymphoid cells on the variable aggressive behavior of PitNET. Regarding the treatment of drug-resistant lactotroph PitNET, proliferative lymphocytes, CD8+ T, and NK cells could represent potentially valuable targets for developing new cancer immunotherapies.
Collapse
Affiliation(s)
- T Elise Potthoff
- Department of Paediatric Haematology and Oncology, University Children's Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Carolin Walter
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
| | - Daniela Jeising
- Department of Paediatric Haematology and Oncology, University Children's Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Daniel Münter
- Department of Paediatric Haematology and Oncology, University Children's Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Archana Verma
- Department of Paediatric Haematology and Oncology, University Children's Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Eric Suero Molina
- Department of Neurosurgery, University Hospital of Münster, 48149, Münster, Germany
| | - Walter Stummer
- Department of Neurosurgery, University Hospital of Münster, 48149, Münster, Germany
| | - Martin Dugas
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
- Institute of Medical Informatics, Heidelberg University Hospital, Heidelberg, Germany
| | - Wolfgang Hartmann
- Division of Translational Pathology, Gerhard Domagk Institute of Pathology, University Hospital Münster, 48149, Münster, Germany
| | - Matthias Dottermusch
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Lea Altendorf
- Department of Paediatric Haematology and Oncology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
- Research Institute Children's Cancer Center, 20251, Hamburg, Germany
| | - Ulrich Schüller
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
- Department of Paediatric Haematology and Oncology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
- Research Institute Children's Cancer Center, 20251, Hamburg, Germany
| | - Sophia Scheuermann
- DFG Cluster of Excellence 2180 'Image-Guided and Functional Instructed Tumor Therapy' (iFIT), University of Tübingen, 72076, Tübingen, Germany
- Department of Pediatric Hematology and Oncology, University Hospital Tübingen, 72076, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site, Tuebingen, Germany
| | - Christian Seitz
- DFG Cluster of Excellence 2180 'Image-Guided and Functional Instructed Tumor Therapy' (iFIT), University of Tübingen, 72076, Tübingen, Germany
- Department of Pediatric Hematology and Oncology, University Hospital Tübingen, 72076, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site, Tuebingen, Germany
| | - Thomas K Albert
- Department of Paediatric Haematology and Oncology, University Children's Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Kornelius Kerl
- Department of Paediatric Haematology and Oncology, University Children's Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.
| |
Collapse
|
74
|
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
|
75
|
Viola MF, Franco Taveras E, Mass E. Developmental programming of tissue-resident macrophages. Front Immunol 2024; 15:1475369. [PMID: 39575254 PMCID: PMC11578957 DOI: 10.3389/fimmu.2024.1475369] [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: 08/03/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024] Open
Abstract
Macrophages are integral components of the innate immune system that colonize organs early in development and persist into adulthood through self-renewal. Their fate, whether they are replaced by monocytes or retain their embryonic origin, depends on tissue type and integrity. Macrophages are influenced by their environment, a phenomenon referred to as developmental programming. This influence extends beyond the local tissue microenvironment and includes soluble factors that can reach the macrophage niche. These factors include metabolites, antibodies, growth factors, and cytokines, which may originate from maternal diet, lifestyle, infections, or other developmental triggers and perturbations. These influences can alter macrophage transcriptional, epigenetic, and metabolic profiles, affecting cell-cell communication and tissue integrity. In addition to their crucial role in tissue immunity, macrophages play vital roles in tissue development and homeostasis. Consequently, developmental programming of these long-lived cells can modulate tissue physiology and pathology throughout life. In this review, we discuss the ontogeny of macrophages, the necessity of developmental programming by the niche for macrophage identity and function, and how developmental perturbations can affect the programming of macrophages and their subtissular niches, thereby influencing disease onset and progression in adulthood. Understanding these effects can inform targeted interventions or preventive strategies against diseases. Finally, understanding the consequences of developmental programming will shed light on how maternal health and disease may impact the well-being of future generations.
Collapse
Affiliation(s)
| | | | - Elvira Mass
- Developmental Biology of the Immune System, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
76
|
Bannazadeh Baghi H, Bayat M, Mehrasa P, Alavi SMA, Lotfalizadeh MH, Memar MY, Taghavi SP, Zarepour F, Hamblin MR, Sadri Nahand J, Hashemian SMR, Mirzaei H. Regulatory role of microRNAs in virus-mediated inflammation. J Inflamm (Lond) 2024; 21:43. [PMID: 39497125 PMCID: PMC11536602 DOI: 10.1186/s12950-024-00417-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 10/21/2024] [Indexed: 11/06/2024] Open
Abstract
Viral infections in humans often cause excessive inflammation. In some viral infections, inflammation can be serious and even fatal, while in other infections it can promote viral clearance. Viruses can escape from the host immune system via regulating inflammatory pathways, thus worsening the illness. MicroRNAs (miRNAs) are tiny non-coding RNA molecules expressed within diverse tissues as well as cells and are engaged in different normal pathological and physiological pathways. Emerging proof suggests that miRNAs can impact innate and adaptive immunity, inflammatory responses, cell invasion, and the progression of viral infections. We discuss some intriguing new findings in the current work, focusing on the impacts of different miRNAs on host inflammatory responses and virus-mediated inflammation. A better understanding of dysregulated miRNAs in viral infections could improve the identification, prevention, and treatment of several serious diseases.
Collapse
Affiliation(s)
- Hossein Bannazadeh Baghi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mobina Bayat
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Plant, Cell and Molecular Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Parisa Mehrasa
- Department of Pathology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Mohammad Yousef Memar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Pouya Taghavi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Zarepour
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Seyed Mohammad Reza Hashemian
- Chronic Respiratory Diseases Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
77
|
Smail MA, Lenz KM. Developmental functions of microglia: Impact of psychosocial and physiological early life stress. Neuropharmacology 2024; 258:110084. [PMID: 39025401 PMCID: PMC12051134 DOI: 10.1016/j.neuropharm.2024.110084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 07/03/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Microglia play numerous important roles in brain development. From early embryonic stages through adolescence, these immune cells influence neuronal genesis and maturation, guide connectivity, and shape brain circuits. They also interact with other glial cells and structures, influencing the brain's supportive microenvironment. While this central role makes microglia essential, it means that early life perturbations to microglia can have widespread effects on brain development, potentially resulting in long-lasting behavioral impairments. Here, we will focus on the effects of early life psychosocial versus physiological stressors in rodent models. Psychosocial stress refers to perceived threats that lead to stress axes activation, including prenatal stress, or chronic postnatal stress, including maternal separation and resource scarcity. Physiological stress refers to physical threats, including maternal immune activation, postnatal infection, and traumatic brain injury. Differing sources of early life stress have varied impacts on microglia, and these effects are moderated by factors such as developmental age, brain region, and sex. Overall, these stressors appear to either 1) upregulate basal microglia numbers and activity throughout the lifespan, while possibly blunting their responsivity to subsequent stressors, or 2) shift the developmental curve of microglia, resulting in differential timing and function, impacting the critical periods they govern. Either could contribute to behavioral dysfunctions that occur after the resolution of early life stress. Exploring how different stressors impact microglia, as well as how multiple stressors interact to alter microglia's developmental functions, could deepen our understanding of how early life stress changes the brain's developmental trajectory. This article is part of the Special Issue on "Microglia".
Collapse
Affiliation(s)
- Marissa A Smail
- Department of Psychology, Ohio State University, Columbus, OH, USA.
| | - Kathryn M Lenz
- Department of Psychology, Ohio State University, Columbus, OH, USA; Department of Neuroscience, Ohio State University, Columbus, OH, USA; Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, Ohio State University, Columbus, OH, USA
| |
Collapse
|
78
|
Couch ACM, Brown AM, Raimundo C, Solomon S, Taylor M, Sichlinger L, Matuleviciute R, Srivastava DP, Vernon AC. Transcriptional and cellular response of hiPSC-derived microglia-neural progenitor co-cultures exposed to IL-6. Brain Behav Immun 2024; 122:27-43. [PMID: 39098436 DOI: 10.1016/j.bbi.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 07/12/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024] Open
Abstract
Elevated interleukin (IL-)6 levels during prenatal development have been linked to increased risk for neurodevelopmental disorders (NDD) in the offspring, but the mechanism remains unclear. Human-induced pluripotent stem cell (hiPSC) models offer a valuable tool to study the effects of IL-6 on features relevant for human neurodevelopment in vitro. We previously reported that hiPSC-derived microglia-like cells (MGLs) respond to IL-6, but neural progenitor cells (NPCs) in monoculture do not. Therefore, we investigated whether co-culturing hiPSC-derived MGLs with NPCs would trigger a cellular response to IL-6 stimulation via secreted factors from the MGLs. Using N=4 donor lines without psychiatric diagnosis, we first confirmed that NPCs can respond to IL-6 through trans-signalling when recombinant IL-6Ra is present, and that this response is dose-dependent. MGLs secreted soluble IL-6R, but at lower levels than found in vivo and below that needed to activate trans-signalling in NPCs. Whilst transcriptomic and secretome analysis confirmed that MGLs undergo substantial transcriptomic changes after IL-6 exposure and subsequently secrete a cytokine milieu, NPCs in co-culture with MGLs exhibited a minimal transcriptional response. Furthermore, there were no significant cell fate-acquisition changes when differentiated into post-mitotic cultures, nor alterations in synaptic densities in mature neurons. These findings highlight the need to investigate if trans-IL-6 signalling to NPCs is a relevant disease mechanism linking prenatal IL-6 exposure to increased risk for psychiatric disorders. Moreover, our findings underscore the importance of establishing more complex in vitro human models with diverse cell types, which may show cell-specific responses to microglia-released cytokines to fully understand how IL-6 exposure may influence human neurodevelopment.
Collapse
Affiliation(s)
- Amalie C M Couch
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| | - Amelia M Brown
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Catarina Raimundo
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Shiden Solomon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Morgan Taylor
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Laura Sichlinger
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Rugile Matuleviciute
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Deepak P Srivastava
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Anthony C Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| |
Collapse
|
79
|
Sypek EI, Tassou A, Collins HY, Huang K, McCallum WM, Bourdillon AT, Barres BA, Bohlen CJ, Scherrer G. Diversity of microglial transcriptional responses during opioid exposure and neuropathic pain. Pain 2024; 165:2615-2628. [PMID: 39073407 PMCID: PMC11474913 DOI: 10.1097/j.pain.0000000000003275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 04/11/2024] [Accepted: 04/14/2024] [Indexed: 07/30/2024]
Abstract
ABSTRACT Microglia take on an altered morphology during chronic opioid treatment. This morphological change is broadly used to identify the activated microglial state associated with opioid side effects, including tolerance and opioid-induced hyperalgesia (OIH). Microglia display similar morphological responses in the spinal cord after peripheral nerve injury (PNI). Consistent with this observation, functional studies have suggested that microglia activated by opioids or PNI engage common molecular mechanisms to induce hypersensitivity. In this article, we conducted deep RNA sequencing (RNA-seq) and morphological analysis of spinal cord microglia in male mice to comprehensively interrogate transcriptional states and mechanistic commonality between multiple models of OIH and PNI. After PNI, we identify an early proliferative transcriptional event across models that precedes the upregulation of histological markers of microglial activation. However, we found no proliferative transcriptional response associated with opioid-induced microglial activation, consistent with histological data, indicating that the number of microglia remains stable during morphine treatment, whereas their morphological response differs from PNI models. Collectively, these results establish the diversity of pain-associated microglial transcriptomic responses and point towards the targeting of distinct insult-specific microglial responses to treat OIH, PNI, or other central nervous system pathologies.
Collapse
Affiliation(s)
- Elizabeth I. Sypek
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, United States
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, United States
- Stanford Neurosciences Institute, Stanford, CA, United States
- Stanford University Neurosciences Graduate Program, Stanford, CA, United States
| | - Adrien Tassou
- Department of Cell Biology and Physiology, UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Hannah Y. Collins
- Department of Neurobiology, Stanford University, Stanford, CA, United States. Bohlen is now with the Department of Neuroscience, Genentech, South San Francisco, CA, United States
| | - Karen Huang
- Department of Cell Biology and Physiology, UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - William M. McCallum
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, United States
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, United States
- Department of Cell Biology and Physiology, UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | | | - Ben A. Barres
- Department of Neurobiology, Stanford University, Stanford, CA, United States. Bohlen is now with the Department of Neuroscience, Genentech, South San Francisco, CA, United States
| | - Christopher J. Bohlen
- Department of Neurobiology, Stanford University, Stanford, CA, United States. Bohlen is now with the Department of Neuroscience, Genentech, South San Francisco, CA, United States
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- New York Stem Cell Foundation—Robertson Investigator Chapel Hill, NC, United States
| |
Collapse
|
80
|
Abdelati Abdelsalam AA, Woods S, Henriquez S, Curran L, Westrop G, Roberts CW. Toxoplasma gondii Infection of BALB/c Mice Perturbs Host Neurochemistry. Parasite Immunol 2024; 46:e13073. [PMID: 39523868 DOI: 10.1111/pim.13073] [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: 08/12/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Toxoplasma gondii infection has been associated with psychoneurological disease in humans and behavioural changes in rodents. However, the mechanisms accounting for this have not been fully described and in some cases could be argued to reflect the severe neuropathology that some mice suffer during infection. Herein we employ a multi-omics approach to extensively examine BALB/c mice that are resistant to toxoplasmic encephalitis. Using a combination of LCMS (liquid chromatography-mass spectrometry) and RNAseq we demonstrate that infection alters the neurochemistry and the transcriptome of the brains of BALB/c mice. Notable changes to tryptophan, purine, arginine, nicotinamide and carnitine metabolism were observed in infected mice and this was accompanied with changes to the levels of a number of transcripts associated with enzymes these metabolic pathways. In addition, changes were seen in transcripts of many immunologically important genes known to contribute to immunity to T. gondii. Changes in the levels of additional transcripts during infection have previously been associated with psychoneurological diseases. The results demonstrate that the BALB/c mouse, with its relatively mild neurological disease, is a useful model for characterising the effects of T. gondii infection on murine neurochemistry. The results also implicate specific biochemical pathways in mediating these changes and should inform further mechanistic studies and suggest therapeutic targets.
Collapse
Affiliation(s)
| | - Stuart Woods
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK
- School of Health & Life Sciences, University of the West of Scotland, South Lanarkshire, UK
| | - Selina Henriquez
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Lucy Curran
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Gareth Westrop
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Craig W Roberts
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
81
|
Xie H, Wu F, Mao J, Wang Y, Zhu J, Zhou X, Hong K, Li B, Qiu X, Wen C. The role of microglia in neurological diseases with involvement of extracellular vesicles. Neurobiol Dis 2024; 202:106700. [PMID: 39401551 DOI: 10.1016/j.nbd.2024.106700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/20/2024] Open
Abstract
As a subset of mononuclear phagocytes in the central nervous system, microglia play a crucial role in immune defense and homeostasis maintenance. Microglia can regulate their states in response to specific signals of health and pathology. Microglia-mediated neuroinflammation is a pathological hallmark of neurodegenerative diseases, neurological damage and neurological tumors, underscoring its key immunoregulatory role in these conditions. Intriguingly, a substantial body of research has indicated that extracellular vesicles can mediate intercellular communication by transporting cargoes from parental cells, a property that is also reflected in microenvironmental signaling networks involving microglia. Based on the microglial characteristics, we briefly outline the biological features of extracellular vesicles and focus on summarizing the integrative role played by microglia in the maintenance of nervous system homeostasis and progression of different neurological diseases. Extracellular vesicles may engage in the homeostasis maintenance and pathological process as a medium of intercellular communication. Here, we aim to provide new insights for further exploration of neurological disease pathogenesis, which may provide theoretical foundations for cell-free therapies.
Collapse
Affiliation(s)
- Haotian Xie
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Feifeng Wu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Jueyi Mao
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yang Wang
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Junquan Zhu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xin Zhou
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Kimsor Hong
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Binbin Li
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xinying Qiu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Chuan Wen
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China.
| |
Collapse
|
82
|
Ma Y, Huang Y, Hu F, Shu K. Lipid metabolic rewiring in glioma‑associated microglia/macrophages (Review). Int J Mol Med 2024; 54:102. [PMID: 39301636 PMCID: PMC11414527 DOI: 10.3892/ijmm.2024.5426] [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: 06/28/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024] Open
Abstract
Gliomas are the most prevailing brain malignancy in both children and adults. Microglia, which are resident in the central nervous system (CNS), are distributed throughout the brain and serve an important role in the immunity of the CNS. Microglial cells exhibit varying phenotypic and metabolic properties during different stages of glioma development, making them a highly dynamic cell population. In particular, glioma‑associated microglia/macrophages (GAMs) can alter their metabolic characteristics and influence malignancies in response to the signals they receive. The significance of macrophage metabolic reprogramming in tumor growth is becoming increasingly acknowledged in recent years. However, to the best of our knowledge, there is currently a scarcity of data from investigations into the lipid metabolic profiles of microglia/macrophages in the glioma setting. Therefore, the present review aims to provide a thorough review of the role that lipid metabolism serves in tumor‑associated macrophages. In addition, it outlines potential targets for therapy based on lipid metabolism. The present review aims to serve as a reference source for future investigations into GAMs.
Collapse
Affiliation(s)
- Yixuan Ma
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
- Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yimin Huang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
- Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Feng Hu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
- Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Kai Shu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
- Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
83
|
Birnbaum EM, Xie L, Serrano P, Rockwell P, Figueiredo-Pereira ME. BT-11 repurposing potential for Alzheimer's disease and insights into its mode of actions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.620882. [PMID: 39553925 PMCID: PMC11565763 DOI: 10.1101/2024.10.29.620882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Neuroinflammation is a key pathological hallmark of Alzheimer's disease (AD). Investigational and FDA approved drugs targeting inflammation already exist, thus drug repurposing for AD is a suitable approach. BT-11 is an investigational drug that reduces inflammation in the gut and improves cognitive function. BT-11 is orally active and binds to lanthionine synthetase C-like 2 (LANCL2), a glutathione-s-transferase, thus potentially reducing oxidative stress. We investigated the effects of BT-11 long-term treatment on the TgF344-AD rat model. BT-11 reduced hippocampal-dependent spatial memory deficits, Aβ plaque load and neuronal loss in males, and mitigated microglia numbers in females. BT-11 treatment led to hippocampal transcriptomic changes in signaling receptor, including G-protein coupled receptor pathways. We detected LANCL2 in hippocampal nuclear and cytoplasmic fractions with potential different post-translational modifications, suggesting distinct functions based on its subcellular localization. LANCL2 was present in oligodendrocytes, showing a role in oligodendrocyte function. To our knowledge, these last two findings have not been reported. Overall, our data suggest that targeting LANCL2 with BT-11 improves cognition and reduces AD-like pathology by potentially modulating G-protein signaling and oligodendrocyte function. Our studies contribute to the field of novel immunomodulatory AD therapeutics, and merit further research on the role of LANCL2 in this disease.
Collapse
|
84
|
Tian G, Bartas K, Hui M, Chen L, Vasquez JJ, Azouz G, Derdeyn P, Manville RW, Ho EL, Fang AS, Li Y, Tyler I, Setola V, Aoto J, Abbott GW, Beier KT. Molecular and circuit determinants in the globus pallidus mediating control of cocaine-induced behavioral plasticity. Neuron 2024; 112:3470-3485.e12. [PMID: 39153478 PMCID: PMC11502257 DOI: 10.1016/j.neuron.2024.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 04/12/2024] [Accepted: 07/22/2024] [Indexed: 08/19/2024]
Abstract
The globus pallidus externus (GPe) is a central component of the basal ganglia circuit that acts as a gatekeeper of cocaine-induced behavioral plasticity. However, the molecular and circuit mechanisms underlying this function are unknown. Here, we show that GPe parvalbumin-positive (GPePV) cells mediate cocaine responses by selectively modulating ventral tegmental area dopamine (VTADA) cells projecting to the dorsomedial striatum (DMS). Interestingly, GPePV cell activity in cocaine-naive mice is correlated with behavioral responses following cocaine, effectively predicting cocaine sensitivity. Expression of the voltage-gated potassium channels KCNQ3 and KCNQ5 that control intrinsic cellular excitability following cocaine was downregulated, contributing to the elevation in GPePV cell excitability. Acutely activating channels containing KCNQ3 and/or KCNQ5 using the small molecule carnosic acid, a key psychoactive component of Salvia rosmarinus (rosemary) extract, reduced GPePV cell excitability and impaired cocaine reward, sensitization, and volitional cocaine intake, indicating its therapeutic potential to counteract psychostimulant use disorder.
Collapse
Affiliation(s)
- Guilian Tian
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Katrina Bartas
- Program in Mathematical, Computational, and Systems Biology, University of California, Irvine, Irvine, CA, USA
| | - May Hui
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Lingxuan Chen
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Jose J Vasquez
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Ghalia Azouz
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Pieter Derdeyn
- Program in Mathematical, Computational, and Systems Biology, University of California, Irvine, Irvine, CA, USA
| | - Rían W Manville
- Bioelectricity Laboratory, Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Erick L Ho
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Amanda S Fang
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Yuan Li
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Isabella Tyler
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Vincent Setola
- Department of Neuroscience, West Virginia University, Morgantown, WV, USA; Department of Behavioral Medicine and Psychiatry, West Virginia University, Morgantown, WV, USA
| | - Jason Aoto
- University of Colorado Anschutz School of Medicine, Department of Pharmacology, Aurora, CO, USA
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Kevin T Beier
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA; Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
85
|
Zeng F, Li Y, Li X, Gu X, Cao Y, Cheng S, Tian H, Mei R, Mei X. Microglia overexpressing brain-derived neurotrophic factor promote vascular repair and functional recovery in mice after spinal cord injury. Neural Regen Res 2024; 21:01300535-990000000-00547. [PMID: 39435607 PMCID: PMC12094574 DOI: 10.4103/nrr.nrr-d-24-00381] [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: 04/07/2024] [Revised: 06/26/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
ABSTRACT Spinal cord injury represents a severe form of central nervous system trauma for which effective treatments remain limited. Microglia is the resident immune cells of the central nervous system, play a critical role in spinal cord injury. Previous studies have shown that microglia can promote neuronal survival by phagocytosing dead cells and debris and by releasing neuroprotective and anti-inflammatory factors. However, excessive activation of microglia can lead to persistent inflammation and contribute to the formation of glial scars, which hinder axonal regeneration. Despite this, the precise role and mechanisms of microglia during the acute phase of spinal cord injury remain controversial and poorly understood. To elucidate the role of microglia in spinal cord injury, we employed the colony-stimulating factor 1 receptor inhibitor PLX5622 to deplete microglia. We observed that sustained depletion of microglia resulted in an expansion of the lesion area, downregulation of brain-derived neurotrophic factor, and impaired functional recovery after spinal cord injury. Next, we generated a transgenic mouse line with conditional overexpression of brain-derived neurotrophic factor specifically in microglia. We found that brain-derived neurotrophic factor overexpression in microglia increased angiogenesis and blood flow following spinal cord injury and facilitated the recovery of hindlimb motor function. Additionally, brain-derived neurotrophic factor overexpression in microglia reduced inflammation and neuronal apoptosis during the acute phase of spinal cord injury. Furthermore, through using specific transgenic mouse lines, TMEM119, and the colony-stimulating factor 1 receptor inhibitor PLX73086, we demonstrated that the neuroprotective effects were predominantly due to brain-derived neurotrophic factor overexpression in microglia rather than macrophages. In conclusion, our findings suggest the critical role of microglia in the formation of protective glial scars. Depleting microglia is detrimental to recovery of spinal cord injury, whereas targeting brain-derived neurotrophic factor overexpression in microglia represents a promising and novel therapeutic strategy to enhance motor function recovery in patients with spinal cord injury.
Collapse
Affiliation(s)
- Fanzhuo Zeng
- Department of Orthopedics, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, China
- Department of Orthopedics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
- Department of Neurobiology, School of Basic Medical Sciences, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yuxin Li
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Xiaoyu Li
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Xinyang Gu
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yue Cao
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Shuai Cheng
- Department of Orthopedics, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - He Tian
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Rongcheng Mei
- Department of Orthopedics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| | - Xifan Mei
- Department of Orthopedics, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, China
| |
Collapse
|
86
|
Tsourmas KI, Butler CA, Kwang NE, Sloane ZR, Dykman KJG, Maloof GO, Prekopa CA, Krattli RP, El-Khatib SM, Swarup V, Acharya MM, Hohsfield LA, Green KN. Myeloid-derived β-hexosaminidase is essential for neuronal health and lysosome function: implications for Sandhoff disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.21.619538. [PMID: 39484433 PMCID: PMC11526954 DOI: 10.1101/2024.10.21.619538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Lysosomal storage disorders (LSDs) are a large disease class involving lysosomal dysfunction, often resulting in neurodegeneration. Sandhoff disease (SD) is an LSD caused by a deficiency in the β subunit of the β-hexosaminidase enzyme (Hexb). Although Hexb expression in the brain is specific to microglia, SD primarily affects neurons. To understand how a microglial gene is involved in maintaining neuronal homeostasis, we demonstrated that β-hexosaminidase is secreted by microglia and integrated into the neuronal lysosomal compartment. To assess therapeutic relevance, we treated SD mice with bone marrow transplant and colony stimulating factor 1 receptor inhibition, which broadly replaced Hexb -/- microglia with Hexb-sufficient cells. This intervention reversed apoptotic gene signatures, improved behavior, restored enzymatic activity and Hexb expression, ameliorated substrate accumulation, and normalized neuronal lysosomal phenotypes. These results underscore the critical role of myeloid-derived β-hexosaminidase in neuronal lysosomal function and establish microglial replacement as a potential LSD therapy.
Collapse
Affiliation(s)
- Kate I. Tsourmas
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Claire A. Butler
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Nellie E. Kwang
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Zachary R. Sloane
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Koby J. G. Dykman
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Ghassan O. Maloof
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Christiana A. Prekopa
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Robert P. Krattli
- Department of Anatomy and Neurobiology; University of California; Irvine, CA 92697; USA
| | - Sanad M. El-Khatib
- Department of Anatomy and Neurobiology; University of California; Irvine, CA 92697; USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Munjal M. Acharya
- Department of Anatomy and Neurobiology; University of California; Irvine, CA 92697; USA
- Department of Radiation Oncology; University of California; Irvine, CA 92697; USA
| | - Lindsay A. Hohsfield
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Kim N. Green
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| |
Collapse
|
87
|
Vecchiarelli HA, Bisht K, Sharma K, Weiser Novak S, Traetta ME, Garcia-Segura ME, St-Pierre MK, Savage JC, Willis C, Picard K, Bordeleau M, Vernoux N, Khakpour M, Garg R, Loewen SM, Murray CJ, Grinberg YY, Faustino J, Halvorson T, Lau V, Pluchino S, Vexler ZS, Carson MJ, Manor U, Peruzzotti-Jametti L, Tremblay MÈ. Dark Microglia Are Abundant in Normal Postnatal Development, where they Remodel Synapses via Phagocytosis and Trogocytosis, and Are Dependent on TREM2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618087. [PMID: 39463930 PMCID: PMC11507757 DOI: 10.1101/2024.10.15.618087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
This study examined dark microglia-a state linked to central nervous system pathology and neurodegeneration-during postnatal development in the mouse ventral hippocampus, finding that dark microglia interact with blood vessels and synapses and perform trogocytosis of pre-synaptic axon terminals. Furthermore, we found that dark microglia in development notably expressed C-type lectin domain family 7 member A (CLEC7a), lipoprotein lipase (LPL) and triggering receptor expressed on myeloid cells 2 (TREM2) and required TREM2, differently from other microglia, suggesting a link between their role in remodeling during development and central nervous system pathology. Together, these results point towards a previously under-appreciated role for dark microglia in synaptic pruning and plasticity during normal postnatal development.
Collapse
|
88
|
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
|
89
|
Sapio MR, Staedtler ES, King DM, Maric D, Jahanipour J, Ghetti A, Jacobson KA, Mannes AJ, Iadarola MJ. Analgesic candidate adenosine A 3 receptors are expressed by perineuronal peripheral macrophages in human dorsal root ganglion and spinal cord microglia. Pain 2024; 165:2323-2343. [PMID: 38691673 PMCID: PMC11408117 DOI: 10.1097/j.pain.0000000000003242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 02/22/2024] [Indexed: 05/03/2024]
Abstract
ABSTRACT Adenosine receptors are a family of purinergic G protein-coupled receptors that are widely distributed in bodily organs and in the peripheral and central nervous systems. Recently, antihyperalgesic actions have been suggested for the adenosine A 3 receptor, and its agonists have been proposed as new neuropathic pain treatments. We hypothesized that these receptors may be expressed in nociceptive primary afferent neurons. However, RNA sequencing across species, eg, rat, mouse, dog, and human, suggests that dorsal root ganglion (DRG) expression of ADORA3 is inconsistent. In rat and mouse, Adora3 shows very weak to no expression in DRG, whereas it is well expressed in human DRG. However, the cell types in human DRG that express ADORA3 have not been delineated. An examination of DRG cell types using in situ hybridization clearly detected ADORA3 transcripts in peripheral macrophages that are in close apposition to the neuronal perikarya but not in peripheral sensory neurons. By contrast, ADORA1 was found primarily in neurons, where it is broadly expressed at low levels. These results suggest that a more complex or indirect mechanism involving modulation of macrophage and/or microglial cells may underlie the potential analgesic action of adenosine A 3 receptor agonism.
Collapse
Affiliation(s)
- Matthew R Sapio
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Ellen S Staedtler
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Diana M King
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Dragan Maric
- National Institute of Neurological Disorders and Stroke, Flow and Imaging Cytometry Core Facility, Bethesda, MD, United States
| | - Jahandar Jahanipour
- National Institute of Neurological Disorders and Stroke, Flow and Imaging Cytometry Core Facility, Bethesda, MD, United States
| | - Andre Ghetti
- AnaBios Corporation, San Diego, CA, United States
| | - Kenneth A Jacobson
- National Institute of Diabetes and Digestive and Kidney Diseases, Molecular Recognition Section, Laboratory of Bioorganic Chemistry, Bethesda, MD, United States
| | - Andrew J Mannes
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Michael J Iadarola
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
90
|
Amo-Aparicio J, Dinarello CA, Lopez-Vales R. Metabolic reprogramming of the inflammatory response in the nervous system: the crossover between inflammation and metabolism. Neural Regen Res 2024; 19:2189-2201. [PMID: 38488552 PMCID: PMC11034585 DOI: 10.4103/1673-5374.391330] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/25/2023] [Accepted: 11/13/2023] [Indexed: 04/24/2024] Open
Abstract
Metabolism is a fundamental process by which biochemicals are broken down to produce energy (catabolism) or used to build macromolecules (anabolism). Metabolism has received renewed attention as a mechanism that generates molecules that modulate multiple cellular responses. This was first identified in cancer cells as the Warburg effect, but it is also present in immunocompetent cells. Studies have revealed a bidirectional influence of cellular metabolism and immune cell function, highlighting the significance of metabolic reprogramming in immune cell activation and effector functions. Metabolic processes such as glycolysis, oxidative phosphorylation, and fatty acid oxidation have been shown to undergo dynamic changes during immune cell response, facilitating the energetic and biosynthetic demands. This review aims to provide a better understanding of the metabolic reprogramming that occurs in different immune cells upon activation, with a special focus on central nervous system disorders. Understanding the metabolic changes of the immune response not only provides insights into the fundamental mechanisms that regulate immune cell function but also opens new approaches for therapeutic strategies aimed at manipulating the immune system.
Collapse
Affiliation(s)
| | | | - Ruben Lopez-Vales
- Institute of Neurosciences, and Department Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Spain
| |
Collapse
|
91
|
Hoffmann A, Miron VE. CNS macrophage contributions to myelin health. Immunol Rev 2024; 327:53-70. [PMID: 39484853 DOI: 10.1111/imr.13416] [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: 11/03/2024]
Abstract
Myelin is the membrane surrounding neuronal axons in the central nervous system (CNS), produced by oligodendrocytes to provide insulation for electrical impulse conduction and trophic/metabolic support. CNS dysfunction occurs following poor development of myelin in infancy, myelin damage in neurological diseases, and impaired regeneration of myelin with disease progression in aging. The lack of approved therapies aimed at supporting myelin health highlights the critical need to identify the cellular and molecular influences on oligodendrocytes. CNS macrophages have been shown to influence the development, maintenance, damage and regeneration of myelin, revealing critical interactions with oligodendrocyte lineage cells. CNS macrophages are comprised of distinct populations, including CNS-resident microglia and cells associated with CNS border regions (the meninges, vasculature, and choroid plexus), in addition to macrophages derived from monocytes infiltrating from the blood. Importantly, the distinct contribution of these macrophage populations to oligodendrocyte lineage responses and myelin health are only just beginning to be uncovered, with the advent of new tools to specifically identify, track, and target macrophage subsets. Here, we summarize the current state of knowledge on the roles of CNS macrophages in myelin health, and recent developments in distinguishing the roles of macrophage populations in development, homeostasis, and disease.
Collapse
Affiliation(s)
- Alana Hoffmann
- BARLO Multiple Sclerosis Centre and Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto, Ontario, Canada
- Department of Immunology, The University of Toronto, Toronto, Ontario, Canada
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Veronique E Miron
- BARLO Multiple Sclerosis Centre and Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto, Ontario, Canada
- Department of Immunology, The University of Toronto, Toronto, Ontario, Canada
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
92
|
Zhao S, Ni K, Xie J, Cheng C, Zhao N, Liu J, Ji W, Wang Q, Zhang P, Liu Y. Exploring the prognostic value of BRMS1 + microglia based on single-cell anoikis regulator patterns in the immunologic microenvironment of GBM. J Neurooncol 2024; 170:101-117. [PMID: 39143438 PMCID: PMC11447114 DOI: 10.1007/s11060-024-04781-5] [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: 04/15/2024] [Accepted: 07/12/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Anoikis is a specialized form of programmed cell death induced by the loss of cell adhesion to the extracellular matrix (ECM). Acquisition of anoikis resistance is a significant marker for cancer cell invasion, metastasis, therapy resistance, and recurrence. Although current research has identified multiple factors that regulate anoikis resistance, the pathological mechanisms of anoikis-mediated tumor microenvironment (TME) in glioblastoma (GBM) remain largely unexplored. METHODS Utilizing single-cell RNA sequencing (scRNA-seq) data and employing non-negative matrix factorization (NMF), we identified and characterized TME cell clusters with distinct anoikis-associated gene signatures. Prognostic and therapeutic response analyses were conducted using TCGA and CGGA datasets to assess the clinical significance of different TME cell clusters. The spatial relationship between BRMS1 + microglia and tumor cells was inferred from spatial transcriptome RNA sequencing (stRNA-seq) data. To simulate the tumor immune microenvironment, co-culture experiments were performed with microglia (HMC3) and GBM cells (U118/U251), and microglia were transfected with a BRMS1 overexpression lentivirus. Western blot or ELISA were used to detect BRMS1, M2 macrophage-specific markers, PI3K/AKT signaling proteins, and apoptosis-related proteins. The proliferation and apoptosis capabilities of tumor cells were evaluated using CCK-8, colony formation, and apoptosis assays, while the invasive and migratory abilities of tumor cells were assessed using Transwell assays. RESULTS NMF-based analysis successfully identified CD8 + T cell and microglia cell clusters with distinct gene signature characteristics. Trajectory analysis, cell communication, and gene regulatory network analyses collectively indicated that anoikis-mediated TME cell clusters can influence tumor cell development through various mechanisms. Notably, BRMS1 + AP-Mic exhibited an M2 macrophage phenotype and had significant cell communication with malignant cells. Moreover, high expression of BRMS1 + AP-Mic in TCGA and CGGA datasets was associated with poorer survival outcomes, indicating its detrimental impact on immunotherapy. Upregulation of BRMS1 in microglia may lead to M2 macrophage polarization, activate the PI3K/AKT signaling pathway through SPP1/CD44-mediated cell interactions, inhibit tumor cell apoptosis, and promote tumor proliferation and invasion. CONCLUSION This pioneering study used NMF-based analysis to reveal the important predictive value of anoikis-regulated TME in GBM for prognosis and immunotherapeutic response. BRMS1 + microglial cells provide a new perspective for a deeper understanding of the immunosuppressive microenvironment of GBM and could serve as a potential therapeutic target in the future.
Collapse
Affiliation(s)
- Songyun Zhao
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center of Nanjing Medical University, Wuxi, China
| | - Kaixiang Ni
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center of Nanjing Medical University, Wuxi, China
| | - Jiaheng Xie
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Chao Cheng
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center of Nanjing Medical University, Wuxi, China
| | - Ning Zhao
- Wuxi Medical Center of Nanjing Medical University, Wuxi, China
| | - Jinhui Liu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Ji
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center of Nanjing Medical University, Wuxi, China
| | - Qi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Pengpeng Zhang
- Department of Lung Cancer Surgery, Tianjin Lung Cancer Center, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.
| | - Yuankun Liu
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China.
- Wuxi Medical Center of Nanjing Medical University, Wuxi, China.
| |
Collapse
|
93
|
Niu X, Zhang Y, Wang Y. Co-culture models for investigating cellular crosstalk in the glioma microenvironment. CANCER PATHOGENESIS AND THERAPY 2024; 2:219-230. [PMID: 39371093 PMCID: PMC11447344 DOI: 10.1016/j.cpt.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 10/08/2024]
Abstract
Glioma is the most prevalent primary malignant tumor in the central nervous system (CNS). It represents a diverse group of brain malignancies characterized by the presence of various cancer cell types as well as an array of noncancerous cells, which together form the intricate glioma tumor microenvironment (TME). Understanding the interactions between glioma cells/glioma stem cells (GSCs) and these noncancerous cells is crucial for exploring the pathogenesis and development of glioma. To invesigate these interactions requires in vitro co-culture models that closely mirror the actual TME in vivo. In this review, we summarize the two- and three-dimensional in vitro co-culture model systems for glioma-TME interactions currently available. Furthermore, we explore common glioma-TME cell interactions based on these models, including interactions of glioma cells/GSCs with endothelial cells/pericytes, microglia/macrophages, T cells, astrocytes, neurons, or other multi-cellular interactions. Together, this review provides an update on the glioma-TME interactions, offering insights into glioma pathogenesis.
Collapse
Affiliation(s)
- Xiaodong Niu
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan Zhang
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuan Wang
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
94
|
Saeki K, Pan R, Lee E, Kurotaki D, Ozato K. IRF8 defines the epigenetic landscape in postnatal microglia, thereby directing their transcriptome programs. Nat Immunol 2024; 25:1928-1942. [PMID: 39313544 DOI: 10.1038/s41590-024-01962-2] [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: 08/14/2023] [Accepted: 08/12/2024] [Indexed: 09/25/2024]
Abstract
Microglia are innate immune cells in the brain. Transcription factor IRF8 (interferon regulatory factor 8) is highly expressed in microglia. However, its role in postnatal microglia development is unknown. We demonstrate that IRF8 binds stepwise to enhancer regions of postnatal microglia along with Sall1 and PU.1, reaching a maximum after day 14. IRF8 binding correlated with a stepwise increase in chromatin accessibility, which preceded the initiation of microglia-specific transcriptome. Constitutive and postnatal Irf8 deletion led to a loss of microglia identity and gain of disease-associated microglia (DAM)-like genes. Combined analysis of single-cell (sc)RNA sequencing and single-cell transposase-accessible chromatin with sequencing (scATAC-seq) revealed a correlation between chromatin accessibility and transcriptome at a single-cell level. IRF8 was also required for microglia-specific DNA methylation patterns. Last, in the 5xFAD model, constitutive and postnatal Irf8 deletion reduced the interaction of microglia with amyloidβ plaques and the size of plaques, lessening neuronal loss. Together, IRF8 sets the epigenetic landscape, which is required for postnatal microglia gene expression.
Collapse
Affiliation(s)
- Keita Saeki
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| | - Richard Pan
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
- Department of Neuroscience, Columbia University, School of Medicine, New York, NY, USA
| | - Eunju Lee
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Daisuke Kurotaki
- Laboratory of Chromatin Organization in Immune Cell Development, International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Keiko Ozato
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
95
|
Chaudhary R, Rehman M, Agarwal V, Kumar A, Kaushik AS, Srivastava S, Srivastava S, Verma R, Rajinikanth PS, Mishra V. Terra incognita of glial cell dynamics in the etiology of leukodystrophies: Broadening disease and therapeutic perspectives. Life Sci 2024; 354:122953. [PMID: 39122110 DOI: 10.1016/j.lfs.2024.122953] [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: 02/05/2024] [Revised: 07/09/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Neuroglial cells, also known as glia, are primarily characterized as auxiliary cells within the central nervous system (CNS). The recent findings have shed light on their significance in numerous physiological processes and their involvement in various neurological disorders. Leukodystrophies encompass an array of rare and hereditary neurodegenerative conditions that were initially characterized by the deficiency, aberration, or degradation of myelin sheath within CNS. The primary cellular populations that experience significant alterations are astrocytes, oligodendrocytes and microglia. These glial cells are either structurally or metabolically impaired due to inherent cellular dysfunction. Alternatively, they may fall victim to the accumulation of harmful by-products resulting from metabolic disturbances. In either situation, the possible replacement of glial cells through the utilization of implanted tissue or stem cell-derived human neural or glial progenitor cells hold great promise as a therapeutic strategy for both the restoration of structural integrity through remyelination and the amelioration of metabolic deficiencies. Various emerging treatment strategies like stem cell therapy, ex-vivo gene therapy, infusion of adeno-associated virus vectors, emerging RNA-based therapies as well as long-term therapies have demonstrated success in pre-clinical studies and show promise for rapid clinical translation. Here, we addressed various leukodystrophies in a comprehensive and detailed manner as well as provide prospective therapeutic interventions that are being considered for clinical trials. Further, we aim to emphasize the crucial role of different glial cells in the pathogenesis of leukodystrophies. By doing so, we hope to advance our understanding of the disease, elucidate underlying mechanisms, and facilitate the development of potential treatment interventions.
Collapse
Affiliation(s)
- Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Mujeeba Rehman
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Vipul Agarwal
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Anand Kumar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Arjun Singh Kaushik
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Siddhi Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Sukriti Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Rajkumar Verma
- University of Connecticut School of Medicine, 200 Academic Way, Farmington, CT 06032, USA
| | - P S Rajinikanth
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Vikas Mishra
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India.
| |
Collapse
|
96
|
Jurcau MC, Jurcau A, Cristian A, Hogea VO, Diaconu RG, Nunkoo VS. Inflammaging and Brain Aging. Int J Mol Sci 2024; 25:10535. [PMID: 39408862 PMCID: PMC11476611 DOI: 10.3390/ijms251910535] [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: 08/29/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Progress made by the medical community in increasing lifespans comes with the costs of increasing the incidence and prevalence of age-related diseases, neurodegenerative ones included. Aging is associated with a series of morphological changes at the tissue and cellular levels in the brain, as well as impairments in signaling pathways and gene transcription, which lead to synaptic dysfunction and cognitive decline. Although we are not able to pinpoint the exact differences between healthy aging and neurodegeneration, research increasingly highlights the involvement of neuroinflammation and chronic systemic inflammation (inflammaging) in the development of age-associated impairments via a series of pathogenic cascades, triggered by dysfunctions of the circadian clock, gut dysbiosis, immunosenescence, or impaired cholinergic signaling. In addition, gender differences in the susceptibility and course of neurodegeneration that appear to be mediated by glial cells emphasize the need for future research in this area and an individualized therapeutic approach. Although rejuvenation research is still in its very early infancy, accumulated knowledge on the various signaling pathways involved in promoting cellular senescence opens the perspective of interfering with these pathways and preventing or delaying senescence.
Collapse
Affiliation(s)
| | - Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, University of Oradea, 410087 Oradea, Romania
| | - Alexander Cristian
- Department of Psycho-Neurosciences and Rehabilitation, University of Oradea, 410087 Oradea, Romania
| | - Vlad Octavian Hogea
- Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | | | | |
Collapse
|
97
|
Stoessel MB, Stowell RD, Lowery RL, Le L, Vu AN, Whitelaw BS, Majewska AK. The effects of P2Y12 loss on microglial gene expression, dynamics, and injury response in the cerebellum and cerebral cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614526. [PMID: 39386439 PMCID: PMC11463386 DOI: 10.1101/2024.09.25.614526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Despite the emerging consensus that microglia are critical to physiological and pathological brain function, it is unclear how microglial roles and their underlying mechanisms differ between brain regions. Microglia throughout the brain express common markers, such as the purinergic receptor P2Y12, that delineate them from peripheral macrophages. P2Y12 is a critical sensor of injury but also contributes to the sensing of neuronal activity and remodeling of synapses, with microglial loss of P2Y12 resulting in behavioral deficits. P2Y12 has largely been studied in cortical microglia, despite the fact that a growing body of evidence suggests that microglia exhibit a high degree of regional specialization. Cerebellar microglia, in particular, exhibit transcriptional, epigenetic, and functional profiles that set them apart from their better studied cortical and hippocampal counterparts. Here, we demonstrate that P2Y12 deficiency does not alter the morphology, distribution, or dynamics of microglia in the cerebellum. In fact, loss of P2Y12 does little to disturb the distinct transcriptomic profiles of cortical and cerebellar microglia. However, unlike in cortex, P2Y12 is not required for a full microglial response to focal injury, suggesting that cerebellar and cortical microglia use different cues to respond to injury. Finally, we show that P2Y12 deficiency impairs cerebellar learning in a delay eyeblink conditioning task, a common test of cerebellar plasticity and circuit function. Our findings suggest not only region-specific roles of microglial P2Y12 signaling in the focal injury response, but also indicate a conserved role for P2Y12 in microglial modulation of plasticity across regions.
Collapse
|
98
|
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
|
99
|
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
|
100
|
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
|