1
|
Li J, Xiao C, Li C, He J. Tissue-resident immune cells: from defining characteristics to roles in diseases. Signal Transduct Target Ther 2025; 10:12. [PMID: 39820040 PMCID: PMC11755756 DOI: 10.1038/s41392-024-02050-5] [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: 06/04/2024] [Revised: 09/28/2024] [Accepted: 11/04/2024] [Indexed: 01/19/2025] Open
Abstract
Tissue-resident immune cells (TRICs) are a highly heterogeneous and plastic subpopulation of immune cells that reside in lymphoid or peripheral tissues without recirculation. These cells are endowed with notably distinct capabilities, setting them apart from their circulating leukocyte counterparts. Many studies demonstrate their complex roles in both health and disease, involving the regulation of homeostasis, protection, and destruction. The advancement of tissue-resolution technologies, such as single-cell sequencing and spatiotemporal omics, provides deeper insights into the cell morphology, characteristic markers, and dynamic transcriptional profiles of TRICs. Currently, the reported TRIC population includes tissue-resident T cells, tissue-resident memory B (BRM) cells, tissue-resident innate lymphocytes, tissue-resident macrophages, tissue-resident neutrophils (TRNs), and tissue-resident mast cells, but unignorably the existence of TRNs is controversial. Previous studies focus on one of them in specific tissues or diseases, however, the origins, developmental trajectories, and intercellular cross-talks of every TRIC type are not fully summarized. In addition, a systemic overview of TRICs in disease progression and the development of parallel therapeutic strategies is lacking. Here, we describe the development and function characteristics of all TRIC types and their major roles in health and diseases. We shed light on how to harness TRICs to offer new therapeutic targets and present burning questions in this field.
Collapse
Affiliation(s)
- Jia Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
2
|
Tacke C, Landgraf P, Dieterich DC, Kröger A. The fate of neuronal synapse homeostasis in aging, infection, and inflammation. Am J Physiol Cell Physiol 2024; 327:C1546-C1563. [PMID: 39495249 DOI: 10.1152/ajpcell.00466.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
Neuroplasticity is the brain's ability to reorganize and modify its neuronal connections in response to environmental stimuli, experiences, learning, and disease processes. This encompasses a variety of mechanisms, including changes in synaptic strength and connectivity, the formation of new synapses, alterations in neuronal structure and function, and the generation of new neurons. Proper functioning of synapses, which facilitate neuron-to-neuron communication, is crucial for brain activity. Neuronal synapse homeostasis, which involves regulating and maintaining synaptic strength and function in the central nervous system (CNS), is vital for this process. Disruptions in synaptic balance, due to factors like inflammation, aging, or infection, can lead to impaired brain function. This review highlights the main aspects and mechanisms underlying synaptic homeostasis, particularly in the context of aging, infection, and inflammation.
Collapse
Affiliation(s)
- Charlotte Tacke
- Institute of Medical Microbiology and Hospital Hygiene, Molecular Microbiology Group, Otto-von-Guericke University, Magdeburg, Germany
| | - Peter Landgraf
- Institute of Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
| | - Daniela C Dieterich
- Institute of Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Andrea Kröger
- Institute of Medical Microbiology and Hospital Hygiene, Molecular Microbiology Group, Otto-von-Guericke University, Magdeburg, Germany
- Helmholtz Center for Infection Research, Innate Immunity and Infection Group, Braunschweig, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
3
|
Niemeyer CS, Merle L, Bubak AN, Baxter BD, Gentile Polese A, Colon-Reyes K, Vang S, Hassell JE, Bruce KD, Nagel MA, Restrepo D. Olfactory and trigeminal routes of HSV-1 CNS infection with regional microglial heterogeneity. J Virol 2024; 98:e0096824. [PMID: 39475273 PMCID: PMC11575344 DOI: 10.1128/jvi.00968-24] [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: 06/03/2024] [Accepted: 09/29/2024] [Indexed: 11/06/2024] Open
Abstract
Herpes simplex virus type 1 (HSV-1) primarily targets the oral and nasal epithelia before establishing latency in the trigeminal ganglion (TG) and other peripheral ganglia. HSV-1 can also infect and become latent in the central nervous system (CNS) independent of latency in the TGs. Recent studies suggest entry to the CNS via two distinct routes: the TG-brainstem connection and olfactory nerve; however, to date, there is no characterization of brain regions targeted during HSV-1 primary infection. Furthermore, the immune response by microglia may also contribute to the heterogeneity between different brain regions. However, the response to HSV-1 by microglia has not been characterized in a region-specific manner. This study investigated the time course of HSV-1 spread within the olfactory epithelium (OE) and CNS following intranasal inoculation and the corresponding macrophage/microglial response in a C57BL/6 mouse model. We found an apical to basal spread of HSV-1 within the OE and underlying tissue accompanied by an inflammatory response of macrophages. OE infection was followed by infection of a small subset of brain regions targeted by the TG in the brainstem and other cranial nerve nuclei, including the vagus and hypoglossal nerve. Furthermore, other brain regions were positive for HSV-1 antigens, such as the locus coeruleus (LC), raphe nucleus (RaN), and hypothalamus while sparing the hippocampus and cortex. Within each brain region, microglia activation also varied widely. These findings provide critical insights into the region-specific dissemination of HSV-1 within the CNS, elucidating potential mechanisms linking viral infection to neurological and neurodegenerative diseases.IMPORTANCEThis study shows how herpes simplex virus type 1 (HSV-1) spreads within the brain after infecting the nasal passages. Our data reveal the distinct pattern of HSV-1 through the brain during a non-encephalitic infection. Furthermore, microglial activation was also temporally and spatially specific, with some regions of the brain having sustained microglial activation even in the absence of viral antigens. Previous reports have identified specific brain regions found to be positive for HSV-1 infection; however, to date, there has not been a concise investigation of the anatomical spread of HSV-1 and the brain regions consistently vulnerable to viral entry and spread. Understanding these region-specific differences in infection and immune response is crucial because it links HSV-1 infection to potential triggers for neurological and neurodegenerative diseases.
Collapse
Affiliation(s)
- Christy S Niemeyer
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Laetitia Merle
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| | - Andrew N Bubak
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - B Dnate' Baxter
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Arianna Gentile Polese
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Katherine Colon-Reyes
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sandy Vang
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - James E Hassell
- Department of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kimberley D Bruce
- Department of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Maria A Nagel
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Diego Restrepo
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
4
|
Niemeyer CS, Merle L, Bubak AN, Dnate' Baxter B, Polese AG, Colon-Reyes K, Vang S, Hassell JE, Bruce KD, Nagel MA, Restrepo D. Olfactory and Trigeminal Routes of HSV-1 CNS Infection with Regional Microglial Heterogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.22.614340. [PMID: 39386674 PMCID: PMC11463476 DOI: 10.1101/2024.09.22.614340] [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
Herpes simplex virus type 1 (HSV-1) primarily targets the oral and nasal epithelia before establishing latency in the trigeminal and other peripheral ganglia (TG). HSV-1 can also infect and go latent in the central nervous system (CNS) independent of latency in the TGs. Recent studies suggest entry to the CNS via two distinct routes: the TG-brainstem connection and olfactory nerve; however, to date, there is no characterization of brain regions targeted during HSV-1 primary infection. Furthermore, the immune response by microglia may also contribute to the heterogeneity between different brain regions. However, the response to HSV-1 by microglia has not been characterized in a region-specific manner. This study investigated the time course of HSV-1 spread within the olfactory epithelium (OE) and CNS following intranasal inoculation and the corresponding macrophage/microglial response in a C57BL/6 mouse model. We found an apical to basal spread of HSV-1 within the OE and underlying tissue accompanied by an inflammatory response of macrophages. OE Infection was followed by infection of a small subset of brain regions targeted by the TG in the brainstem, as well as other cranial nerve nuclei, including the vagus and hypoglossal nerve. Furthermore, other brain regions were positive for HSV-1 antigens, such as the locus coeruleus (LC), raphe nucleus (RaN), and hypothalamus, while sparing the hippocampus and cortex. Within each brain region, microglia activation also varied widely. These findings provide critical insights into the region-specific dissemination of HSV-1 within the CNS, elucidating potential mechanisms linking viral infection to neurological and neurodegenerative diseases. Importance This study sheds light on how herpes simplex virus type 1 (HSV-1) spreads within the brain after infecting the nasal passages. Our data reveals the distinct pattern of HSV-1 through the brain during a non-encephalitic infection. Furthermore, microglial activation was also temporally and spatially specific, with some regions of the brain having sustained microglial activation even in the absence of viral antigen. Previous reports have identified specific regions of the brain found to be positive for HSV-1 infection; however, to date, there has not been a concise investigation of the anatomical spread of HSV-1 and the regions of the brain consistently vulnerable to viral entry and spread. Understanding these region-specific differences in infection and immune response is crucial because it links HSV-1 infection to potential triggers for neurological and neurodegenerative diseases.
Collapse
|
5
|
Liu J, Chen X, Liu J, Zhang H, Lu W. HSV-1 immune escapes in microglia by down-regulating GM130 to inhibit TLR3-mediated innate immune responses. Virol J 2024; 21:219. [PMID: 39285274 PMCID: PMC11404012 DOI: 10.1186/s12985-024-02492-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 09/06/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND To investigate the mechanism of Golgi matrix protein 130(GM130) regulating the antiviral immune response of TLR3 after herpes simplex virus type 1(HSV-1) infection of microglia cells. We explored the regulatory effects of berberine on the immune response mediated by GM130 and TLR3. METHODS An in vitro model of HSV-1 infection was established by infecting BV2 cells with HSV-1. RESULTS Compared to the uninfected group, the Golgi apparatus (GA) fragmentation and GM130 decreased after HSV-1 infection; TLR3 increased at 6 h and began to decrease at 12 h after HSV-1 infection; the secretion of interferon-beta(IFN-β), tumour necrosis factor alpha(TNF-α), and interleukin-6(IL-6) increased after infection. Knockdown of GM130 aggravated fragmentation of the GA and caused TLR3 to further decrease, and the virus titer also increased significantly. GM130 knockdown inhibits the increase in TLR3 and inflammatory factors induced by TLR3 agonists and increases the viral titer. Overexpression of GM130 alleviated fragmentation of the GA induced by HSV-1, partially restored the levels of TLR3, and reduced viral titers. GM130 overexpression reversed the reduction in TLR3 and inflammatory cytokine levels induced by TLR3 inhibitors. Therefore, the decrease in GM130 levels caused by HSV-1 infection leads to increased viral replication by inhibiting TLR3-mediated innate immunity. Berberine can protect the GA and reverse the downregulation of GM130, as well as the downregulation of TLR3 and its downstream factors after HSV-1 infection, reducing the virus titer. CONCLUSIONS In microglia, one mechanism of HSV-1 immune escape is disruption of the GM130/TLR3 pathway. Berberine protects the GA and enhances TLR3-mediated antiviral immune responses.
Collapse
Affiliation(s)
- Jia Liu
- Department of Neurology, Second Xiangya Hospital, Central South University, No. 139, Renmin Middle Road, Changsha, Hunan, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, No. 139, Renmin Middle Road, Changsha, Hunan, China
| | - Xiqian Chen
- Department of Neurology, Second Xiangya Hospital, Central South University, No. 139, Renmin Middle Road, Changsha, Hunan, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, No. 139, Renmin Middle Road, Changsha, Hunan, China
| | - Junxian Liu
- Department of Neurology, Second Xiangya Hospital, Central South University, No. 139, Renmin Middle Road, Changsha, Hunan, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, No. 139, Renmin Middle Road, Changsha, Hunan, China
| | - Hainan Zhang
- Department of Neurology, Second Xiangya Hospital, Central South University, No. 139, Renmin Middle Road, Changsha, Hunan, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, No. 139, Renmin Middle Road, Changsha, Hunan, China
| | - Wei Lu
- Department of Neurology, Second Xiangya Hospital, Central South University, No. 139, Renmin Middle Road, Changsha, Hunan, China.
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, No. 139, Renmin Middle Road, Changsha, Hunan, China.
| |
Collapse
|
6
|
Pavlou A, Mulenge F, Gern OL, Busker LM, Greimel E, Waltl I, Kalinke U. Orchestration of antiviral responses within the infected central nervous system. Cell Mol Immunol 2024; 21:943-958. [PMID: 38997413 PMCID: PMC11364666 DOI: 10.1038/s41423-024-01181-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/05/2024] [Indexed: 07/14/2024] Open
Abstract
Many newly emerging and re-emerging viruses have neuroinvasive potential, underscoring viral encephalitis as a global research priority. Upon entry of the virus into the CNS, severe neurological life-threatening conditions may manifest that are associated with high morbidity and mortality. The currently available therapeutic arsenal against viral encephalitis is rather limited, emphasizing the need to better understand the conditions of local antiviral immunity within the infected CNS. In this review, we discuss new insights into the pathophysiology of viral encephalitis, with a focus on myeloid cells and CD8+ T cells, which critically contribute to protection against viral CNS infection. By illuminating the prerequisites of myeloid and T cell activation, discussing new discoveries regarding their transcriptional signatures, and dissecting the mechanisms of their recruitment to sites of viral replication within the CNS, we aim to further delineate the complexity of antiviral responses within the infected CNS. Moreover, we summarize the current knowledge in the field of virus infection and neurodegeneration and discuss the potential links of some neurotropic viruses with certain pathological hallmarks observed in neurodegeneration.
Collapse
Affiliation(s)
- Andreas Pavlou
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Felix Mulenge
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Olivia Luise Gern
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Lena Mareike Busker
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, 30559, Hannover, Germany
| | - Elisabeth Greimel
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Inken Waltl
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany.
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
7
|
Krzyzowska M, Patrycy M, Chodkowski M, Janicka M, Kowalczyk A, Skulska K, Thörn K, Eriksson K. Fas/FasL-Mediated Apoptosis and Inflammation Contribute to Recovery from HSV-2-Mediated Spinal Cord Infection. Viruses 2024; 16:1363. [PMID: 39339840 PMCID: PMC11436029 DOI: 10.3390/v16091363] [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: 07/21/2024] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
Herpes simplex virus type 2 (HSV-2) is a sexually transmitted pathogen that causes a persistent infection in sensory ganglia. The infection manifests itself as genital herpes but in rare cases it can cause meningitis. In this study, we used a murine model of HSV-2 meningitis to show that Fas and FasL are induced within the CNS upon HSV-2 infection, both on resident microglia and astrocytes and on infiltrating monocytes and lymphocytes. Mice lacking Fas or FasL had a more severe disease development with significantly higher morbidity, mortality, and an overall higher CNS viral load. In parallel, these Fas/FasL-deficient mice showed a severely impaired infection-induced CNS inflammatory response with lower levels of infiltrating CD4+ T-cells, lower levels of Th1 cytokines and chemokines, and a shift in the balance between M1 and M2 microglia/monocytes. In vitro, we confirmed that Fas and FasL is required for the induction of leucocyte apoptosis, but also show that the Fas/FasL pathway is required for adequate cytokine and chemokine production by glial cells. In summary, our data show that the Fas/FasL cell death receptor pathway is an important defense mechanism in the spinal cord as it down-regulates HSV-2-induced inflammation while at the same time promoting adequate anti-viral immune responses against infection.
Collapse
Affiliation(s)
- Malgorzata Krzyzowska
- Military Institute of Hygiene and Epidemiology, 01-163 Warsaw, Poland; (M.P.); (M.C.); (M.J.)
| | - Magdalena Patrycy
- Military Institute of Hygiene and Epidemiology, 01-163 Warsaw, Poland; (M.P.); (M.C.); (M.J.)
| | - Marcin Chodkowski
- Military Institute of Hygiene and Epidemiology, 01-163 Warsaw, Poland; (M.P.); (M.C.); (M.J.)
| | - Martyna Janicka
- Military Institute of Hygiene and Epidemiology, 01-163 Warsaw, Poland; (M.P.); (M.C.); (M.J.)
| | - Andrzej Kowalczyk
- PORT Polish Center for Technology Development, 54-066 Wroclaw, Poland; (A.K.); (K.S.)
| | - Katarzyna Skulska
- PORT Polish Center for Technology Development, 54-066 Wroclaw, Poland; (A.K.); (K.S.)
| | - Karolina Thörn
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (K.T.); (K.E.)
| | - Kristina Eriksson
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (K.T.); (K.E.)
| |
Collapse
|
8
|
Mulenge F, Gern OL, Busker LM, Aringo A, Ghita L, Waltl I, Pavlou A, Kalinke U. Transcriptomic analysis unveils bona fide molecular signatures of microglia under conditions of homeostasis and viral encephalitis. J Neuroinflammation 2024; 21:203. [PMID: 39153993 PMCID: PMC11330067 DOI: 10.1186/s12974-024-03197-2] [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: 04/24/2024] [Accepted: 08/06/2024] [Indexed: 08/19/2024] Open
Abstract
Microglia serve as a front-line defense against neuroinvasive viral infection, however, determination of their actual transcriptional profiles under conditions of health and disease is challenging. Here, we used various experimental approaches to delineate the transcriptional landscape of microglia during viral infection. Intriguingly, multiple activation genes were found to be artificially induced in sorted microglia and we demonstrated that shear stress encountered during cell sorting was one of the key inducers. Post-hoc analysis revealed that publicly available large-scale single-cell RNA sequencing datasets were significantly tainted by aberrant signatures that are associated with cell sorting. By exploiting the ribosomal tagging approach, we developed a strategy to enrich microglia-specific transcripts by comparing immunoprecipitated RNA with total RNA. Such enriched transcripts were instrumental in defining bona fide signatures of microglia under conditions of health and virus infection. These unified microglial signatures may serve as a benchmark to retrospectively assess ex vivo artefacts from available atlases. Leveraging the microglial translatome, we found enrichment of genes implicated in T-cell activation and cytokine production during the course of VSV infection. These data linked microglia with T-cell re-stimulation and further underscored that microglia are involved in shaping antiviral T-cell responses in the brain. Collectively, our study defines the transcriptional landscape of microglia under steady state and during viral encephalitis and highlights cellular interactions between microglia and T cells that contribute to the control of virus dissemination.
Collapse
Affiliation(s)
- Felix Mulenge
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, a joint venture between The Helmholtz-Centre for Infection Research, Hannover Medical School, TWINCORE, Feodor-Lynen-Str. 7, 30625, Hannover, Germany
| | - Olivia Luise Gern
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, a joint venture between The Helmholtz-Centre for Infection Research, Hannover Medical School, TWINCORE, Feodor-Lynen-Str. 7, 30625, Hannover, Germany
| | - Lena Mareike Busker
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, a joint venture between The Helmholtz-Centre for Infection Research, Hannover Medical School, TWINCORE, Feodor-Lynen-Str. 7, 30625, Hannover, Germany
- Department of Pathology, University of Veterinary Medicine Hannover, 30559, Foundation, Hannover, Germany
| | - Angela Aringo
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, a joint venture between The Helmholtz-Centre for Infection Research, Hannover Medical School, TWINCORE, Feodor-Lynen-Str. 7, 30625, Hannover, Germany
| | - Luca Ghita
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, a joint venture between The Helmholtz-Centre for Infection Research, Hannover Medical School, TWINCORE, Feodor-Lynen-Str. 7, 30625, Hannover, Germany
- , Genentech, South San Francisco, CA, 94080, USA
| | - Inken Waltl
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, a joint venture between The Helmholtz-Centre for Infection Research, Hannover Medical School, TWINCORE, Feodor-Lynen-Str. 7, 30625, Hannover, Germany
| | - Andreas Pavlou
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, a joint venture between The Helmholtz-Centre for Infection Research, Hannover Medical School, TWINCORE, Feodor-Lynen-Str. 7, 30625, Hannover, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, a joint venture between The Helmholtz-Centre for Infection Research, Hannover Medical School, TWINCORE, Feodor-Lynen-Str. 7, 30625, Hannover, Germany.
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
9
|
Fruhwürth S, Zetterberg H, Paludan SR. Microglia and amyloid plaque formation in Alzheimer's disease - Evidence, possible mechanisms, and future challenges. J Neuroimmunol 2024; 390:578342. [PMID: 38640827 DOI: 10.1016/j.jneuroim.2024.578342] [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: 02/01/2024] [Revised: 03/21/2024] [Accepted: 04/03/2024] [Indexed: 04/21/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by cognitive decline that severely affects patients and their families. Genetic and environmental risk factors, such as viral infections, synergize to accelerate the aging-associated neurodegeneration. Genetic risk factors for late-onset AD (LOAD), which accounts for most AD cases, are predominantly implicated in microglial and immune cell functions. As such, microglia play a major role in formation of amyloid beta (Aβ) plaques, the major pathological hallmark of AD. This review aims to provide an overview of the current knowledge regarding the role of microglia in Aβ plaque formation, as well as their impact on morphological and functional diversity of Aβ plaques. Based on this discussion, we seek to identify challenges and opportunities in this field with potential therapeutic implications.
Collapse
Affiliation(s)
- Stefanie Fruhwürth
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, Institute of Neurology, University College London Queen Square, London, UK; UK Dementia Research Institute at UCL, London, UK; Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, USA
| | - Søren R Paludan
- Department of Rheumatology and Inflammatory Research, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
10
|
Weyer MP, Strehle J, Schäfer MKE, Tegeder I. Repurposing of pexidartinib for microglia depletion and renewal. Pharmacol Ther 2024; 253:108565. [PMID: 38052308 DOI: 10.1016/j.pharmthera.2023.108565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/07/2023]
Abstract
Pexidartinib (PLX3397) is a small molecule receptor tyrosine kinase inhibitor of colony stimulating factor 1 receptor (CSF1R) with moderate selectivity over other members of the platelet derived growth factor receptor family. It is approved for treatment of tenosynovial giant cell tumors (TGCT). CSF1R is highly expressed by microglia, which are macrophages of the central nervous system (CNS) that defend the CNS against injury and pathogens and contribute to synapse development and plasticity. Challenged by pathogens, apoptotic cells, debris, or inflammatory molecules they adopt a responsive state to propagate the inflammation and eventually return to a homeostatic state. The phenotypic switch may fail, and disease-associated microglia contribute to the pathophysiology in neurodegenerative or neuropsychiatric diseases or long-lasting detrimental brain inflammation after brain, spinal cord or nerve injury or ischemia/hemorrhage. Microglia also contribute to the growth permissive tumor microenvironment of glioblastoma (GBM). In rodents, continuous treatment for 1-2 weeks via pexidartinib food pellets leads to a depletion of microglia and subsequent repopulation from the remaining fraction, which is aided by peripheral monocytes that search empty niches for engraftment. The putative therapeutic benefit of such microglia depletion or forced renewal has been assessed in almost any rodent model of CNS disease or injury or GBM with heterogeneous outcomes, but a tendency of partial beneficial effects. So far, microglia monitoring e.g. via positron emission imaging is not standard of care for patients receiving Pexidartinib (e.g. for TGCT), so that the depletion and repopulation efficiency in humans is still largely unknown. Considering the virtuous functions of microglia, continuous depletion is likely no therapeutic option but short-lasting transient partial depletion to stimulate microglia renewal or replace microglia in genetic disease in combination with e.g. stem cell transplantation or as part of a multimodal concept in treatment of glioblastoma appears feasible. The present review provides an overview of the preclinical evidence pro and contra microglia depletion as a therapeutic approach.
Collapse
Affiliation(s)
- Marc-Philipp Weyer
- Institute of Clinical Pharmacology, Goethe-University Frankfurt, Faculty of Medicine, Frankfurt, Germany
| | - Jenny Strehle
- Department of Anesthesiology, University Medical Center Johannes Gutenberg-University Mainz, Germany
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center Johannes Gutenberg-University Mainz, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Goethe-University Frankfurt, Faculty of Medicine, Frankfurt, Germany.
| |
Collapse
|
11
|
Awogbindin I, Wanklin M, Verkhratsky A, Tremblay MÈ. Microglia in Neurodegenerative Diseases. ADVANCES IN NEUROBIOLOGY 2024; 37:497-512. [PMID: 39207709 DOI: 10.1007/978-3-031-55529-9_27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Neurodegenerative diseases are manifested by a progressive death of neural cells, resulting in the deterioration of central nervous system (CNS) functions, ultimately leading to specific behavioural and cognitive symptoms associated with affected brain regions. Several neurodegenerative disorders are caused by genetic variants or mutations, although the majority of cases are sporadic and linked to various environmental risk factors, with yet an unknown aetiology. Neuroglial changes are fundamental and often lead to the pathophysiology of neurodegenerative diseases. In particular, microglial cells, which are essential for maintaining CNS health, become compromised in their physiological functions with the exposure to environmental risk factors, genetic variants or mutations, as well as disease pathology. In this chapter, we cover the contribution of neuroglia, especially microglia, to several neurodegenerative diseases, including Nasu-Hakola disease, Parkinson's disease, amyotrophic lateral sclerosis, Alzheimer's disease, Huntington's disease, infectious disease-associated neurodegeneration, and metal-precipitated neurodegeneration. Future research perspectives for the field pertaining to the therapeutic targeting of microglia across these disease conditions are also discussed.
Collapse
Affiliation(s)
- Ifeoluwa Awogbindin
- Department of Biochemistry, Neuroimmunology Group, Molecular Drug Metabolism and Toxicology Laboratory, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, Canada
- Institute on Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada
| | - Michael Wanklin
- Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, Canada
| | - Alexei Verkhratsky
- Faculty of Life Sciences, The University of Manchester, Manchester, UK.
- Department of Neurosciences, University of the Basque Country, Leioa, Bizkaia, Spain.
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China.
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada.
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| | - Marie-Ève Tremblay
- Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, Canada.
- Institute on Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada.
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
- Department of Molecular Medicine, Université Laval, Pavillon Ferdinand-Vandry, Québec City, QC, Canada.
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Life Sciences Center, Vancouver, BC, Canada.
| |
Collapse
|
12
|
Fruhwürth S, Reinert LS, Öberg C, Sakr M, Henricsson M, Zetterberg H, Paludan SR. TREM2 is down-regulated by HSV1 in microglia and involved in antiviral defense in the brain. SCIENCE ADVANCES 2023; 9:eadf5808. [PMID: 37595041 PMCID: PMC10438464 DOI: 10.1126/sciadv.adf5808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 07/19/2023] [Indexed: 08/20/2023]
Abstract
Immunological control of viral infections in the brain exerts immediate protection and also long-term maintenance of brain integrity. Microglia are important for antiviral defense in the brain. Here, we report that herpes simplex virus type 1 (HSV1) infection of human induced pluripotent stem cell (hiPSC)-derived microglia down-regulates expression of genes in the TREM2 pathway. TREM2 was found to be important for virus-induced IFNB induction through the DNA-sensing cGAS-STING pathway in microglia and for phagocytosis of HSV1-infected neurons. Consequently, TREM2 depletion increased susceptibility to HSV1 infection in human microglia-neuron cocultures and in the mouse brain. TREM2 augmented STING signaling and activation of downstream targets TBK1 and IRF3. Thus, TREM2 is important for the antiviral immune response in microglia. Since TREM2 loss-of-function mutations and HSV1 serological status are both linked to Alzheimer's disease, this work poses the question whether genetic or virus-induced alterations of TREM2 activity predispose to post-infection neurological pathologies.
Collapse
Affiliation(s)
- Stefanie Fruhwürth
- Department of Rheumatology and Inflammatory Research, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Line S. Reinert
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Carl Öberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Marcelina Sakr
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Marcus Henricsson
- Biomarker Discovery and Development, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Søren R. Paludan
- Department of Rheumatology and Inflammatory Research, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
13
|
McMillan RE, Wang E, Carlin AF, Coufal NG. Human microglial models to study host-virus interactions. Exp Neurol 2023; 363:114375. [PMID: 36907350 PMCID: PMC10521930 DOI: 10.1016/j.expneurol.2023.114375] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/13/2023] [Accepted: 03/02/2023] [Indexed: 03/14/2023]
Abstract
Microglia, the resident macrophage of the central nervous system, are increasingly recognized as contributing to diverse aspects of human development, health, and disease. In recent years, numerous studies in both mouse and human models have identified microglia as a "double edged sword" in the progression of neurotropic viral infections: protecting against viral replication and cell death in some contexts, while acting as viral reservoirs and promoting excess cellular stress and cytotoxicity in others. It is imperative to understand the diversity of human microglial responses in order to therapeutically modulate them; however, modeling human microglia has been historically challenging due to significant interspecies differences in innate immunity and rapid transformation upon in vitro culture. In this review, we discuss the contribution of microglia to the neuropathogenesis of key neurotropic viral infections: human immunodeficiency virus 1 (HIV-1), Zika virus (ZIKV), Japanese encephalitis virus (JEV), West Nile virus (WNV), Herpes simplex virus (HSV), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We pay special attention to recent work with human stem cell-derived microglia and propose strategies to leverage these powerful models to further uncover species- and disease-specific microglial responses and novel therapeutic interventions for neurotropic viral infections.
Collapse
Affiliation(s)
- Rachel E McMillan
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, United States of America; Department of Pathology and Medicine, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States of America
| | - Ellen Wang
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States of America; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92093, United States of America
| | - Aaron F Carlin
- Department of Pathology and Medicine, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States of America.
| | - Nicole G Coufal
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States of America; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92093, United States of America.
| |
Collapse
|
14
|
Spiteri AG, Wishart CL, Ni D, Viengkhou B, Macia L, Hofer MJ, King NJC. Temporal tracking of microglial and monocyte single-cell transcriptomics in lethal flavivirus infection. Acta Neuropathol Commun 2023; 11:60. [PMID: 37016414 PMCID: PMC10074823 DOI: 10.1186/s40478-023-01547-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/08/2023] [Indexed: 04/06/2023] Open
Abstract
As the resident parenchymal myeloid population in the central nervous system (CNS), microglia are strategically positioned to respond to neurotropic virus invasion and have been implicated in promoting both disease resolution and progression in the acute and post-infectious phase of virus encephalitis. In a mouse model of West Nile virus encephalitis (WNE), infection of the CNS results in recruitment of large numbers of peripheral immune cells into the brain, the majority being nitric oxide (NO)-producing Ly6Chi inflammatory monocyte-derived cells (MCs). In this model, these cells enhance immunopathology and mortality. However, the contribution of microglia to this response is currently undefined. Here we used a combination of experimental tools, including single-cell RNA sequencing (scRNA-seq), microglia and MC depletion reagents, high-dimensional spectral cytometry and computational algorithms to dissect the differential contribution of microglia and MCs to the anti-viral immune response in severe neuroinflammation seen in WNE. Intriguingly, analysis of scRNA-seq data revealed 6 unique microglia and 3 unique MC clusters that were predominantly timepoint-specific, demonstrating substantial transcriptional adaptation with disease progression over the course of WNE. While microglia and MC adopted unique gene expression profiles, gene ontology enrichment analysis, coupled with microglia and MC depletion studies, demonstrated a role for both of these cells in the trafficking of peripheral immune cells into the CNS, T cell responses and viral clearance. Over the course of infection, microglia transitioned from a homeostatic to an anti-viral and then into an immune cell-recruiting phenotype. Conversely, MC adopted antigen-presenting, immune cell-recruiting and NO-producing phenotypes, which all had anti-viral function. Overall, this study defines for the first time the single-cell transcriptomic responses of microglia and MCs over the course of WNE, demonstrating both protective and pathological roles of these cells that could potentially be targeted for differential therapeutic intervention to dampen immune-mediated pathology, while maintaining viral clearance functions.
Collapse
Affiliation(s)
- Alanna G Spiteri
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, NSW, 2006, Australia
- Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Claire L Wishart
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, NSW, 2006, Australia
- Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Duan Ni
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
- Chronic Diseases Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Barney Viengkhou
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Laurence Macia
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
- Chronic Diseases Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Markus J Hofer
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
- The University of Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Nicholas J C King
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia.
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, NSW, 2006, Australia.
- Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, NSW, 2006, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia.
- The University of Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, NSW, 2006, Australia.
- Sydney Nano, The University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
15
|
Patrycy M, Chodkowski M, Krzyzowska M. Role of Microglia in Herpesvirus-Related Neuroinflammation and Neurodegeneration. Pathogens 2022; 11:pathogens11070809. [PMID: 35890053 PMCID: PMC9324537 DOI: 10.3390/pathogens11070809] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/15/2022] [Accepted: 07/17/2022] [Indexed: 02/04/2023] Open
Abstract
Neuroinflammation is defined as an inflammatory state within the central nervous system (CNS). Microglia conprise the resident tissue macrophages of the neuronal tissue. Upon viral infection of the CNS, microglia become activated and start to produce inflammatory mediators important for clearance of the virus, but an excessive neuroinflammation can harm nearby neuronal cells. Herpesviruses express several molecular mechanisms, which can modulate apoptosis of infected neurons, astrocytes and microglia but also divert immune response initiated by the infected cells. In this review we also describe the link between virus-related neuroinflammation, and development of neurodegenerative diseases.
Collapse
|
16
|
Li F, Wang Y, Song X, Wang Z, Jia J, Qing S, Huang L, Wang Y, Wang S, Ren Z, Zheng K, Wang Y. The intestinal microbial metabolite nicotinamide n-oxide prevents herpes simplex encephalitis via activating mitophagy in microglia. Gut Microbes 2022; 14:2096989. [PMID: 35793266 PMCID: PMC9262364 DOI: 10.1080/19490976.2022.2096989] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Herpes simplex encephalitis (HSE), a complication of herpes simplex virus type I (HSV-1) infection causes neurological disorder or even death in immunocompromised adults and newborns. However, the intrinsic factors controlling the HSE outcome remain unclear. Here, we show that HSE mice exhibit gut microbiota dysbiosis and altered metabolite configuration and tryptophan-nicotinamide metabolism. HSV-1 neurotropic infection activated microglia, with changed immune properties and cell numbers, to stimulate antiviral immune response and contribute substantially to HSE. In addition, depletion of gut microbiota by oral antibiotics (ABX)-treatment triggered the hyper-activation of microglia, which in turn enhanced inflammatory immune response, and cytokine production, resulting in aggregated viral burden and HSE pathology. Furthermore, exogenous administration of nicotinamide n-oxide (NAMO), an oxidative product of nicotinamide derived from gut microbiota, to ABX-treated or untreated HSE mice significantly diminished microglia-mediated proinflammatory response and limited HSV-1 infection in CNS. Mechanistic study revealed that HSV-1 activates microglia by increasing mitochondrial damage via defective mitophagy, whereas microbial metabolite NAMO restores NAD+-dependent mitophagy to inhibit microglia activation and HSE progression. NAMO also prevented neuronal cell death triggered by HSV-1 infection or microglia-mediated microenvironmental toxicity. Finally, we show that NAMO is mainly generated by neomycin-sensitive bacteria, especially Lactobacillus_gasseri and Lactobacillus_reuteri. Together, these data demonstrate that gut microbial metabolites act as intrinsic restrictive factors against HSE progression via regulating mitophagy in microglia, implying further exploration of bacterial or nutritional approaches for treating neurotropic virus-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Feng Li
- Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China,Infectious Diseases Institute, Guangzhou Eighth People’s Hospital, Guangdong, China
| | - Yiliang Wang
- Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Xiaowei Song
- Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Zhaoyang Wang
- Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Jiaoyan Jia
- Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Shurong Qing
- Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Lianzhou Huang
- Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Yuan Wang
- Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Shuai Wang
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhe Ren
- Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China,Zhe Ren Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou510632, China
| | - Kai Zheng
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, Guangdong, China,Kai Zheng School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, Guangdong, China
| | - Yifei Wang
- Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China,CONTACT Yifei Wang
| |
Collapse
|
17
|
Krzyzowska M, Jarneborn A, Thorn K, Eriksson K, Jin T. Tofacitinib Treatment in Primary Herpes Simplex Encephalitis Interferes With Antiviral Response. J Infect Dis 2022; 225:1545-1553. [PMID: 35217873 PMCID: PMC9635063 DOI: 10.1093/infdis/jiac040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 01/31/2022] [Indexed: 10/29/2023] Open
Abstract
Tofacitinib, a Janus kinase inhibitor, is a novel immunosuppressive drug for treatment of rheumatoid arthritis. Herpes simplex virus type 1 (HSV-1) may cause encephalitis during primary infection or following reactivation from a latent state. Long-term tofacitinib treatment may increase the risk of this life-threatening condition. The aim of this study was to investigate the effect of tofacitinib on HSV-1 primary infection using a mouse model. Mice pretreated with tofacitinib were intranasally infected with a clinical strain of HSV-1 and monitored for infection severity and antiviral response. Tofacitinib treatment of HSV-1 primary infection resulted in increased viral loads and worsened clinical outcome. Furthermore, tofacitinib promoted M2 anti-inflammatory phenotype of microglia and infiltrating monocytes, as well as inhibited production of inflammatory and antiviral cytokines by macrophages in vitro. Our findings show that treatment with tofacitinib increases severity of herpes simplex encephalitis in mice, by impairing antiviral response induced by monocytes and microglia.
Collapse
Affiliation(s)
- Malgorzata Krzyzowska
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| | - Anders Jarneborn
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Karolina Thorn
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kristina Eriksson
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
18
|
Stables J, Green EK, Sehgal A, Patkar OL, Keshvari S, Taylor I, Ashcroft ME, Grabert K, Wollscheid-Lengeling E, Szymkowiak S, McColl BW, Adamson A, Humphreys NE, Mueller W, Starobova H, Vetter I, Shabestari SK, Blurton-Jones MM, Summers KM, Irvine KM, Pridans C, Hume DA. A kinase-dead Csf1r mutation associated with adult-onset leukoencephalopathy has a dominant inhibitory impact on CSF1R signalling. Development 2022; 149:274819. [PMID: 35333324 PMCID: PMC9002114 DOI: 10.1242/dev.200237] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 02/04/2022] [Indexed: 12/21/2022]
Abstract
Amino acid substitutions in the kinase domain of the human CSF1R gene are associated with autosomal dominant adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP). To model the human disease, we created a disease-associated mutation (pGlu631Lys; E631K) in the mouse Csf1r locus. Homozygous mutation (Csf1rE631K/E631K) phenocopied the Csf1r knockout, with prenatal mortality or severe postnatal growth retardation and hydrocephalus. Heterozygous mutation delayed the postnatal expansion of tissue macrophage populations in most organs. Bone marrow cells from Csf1rE631K/+mice were resistant to CSF1 stimulation in vitro, and Csf1rE631K/+ mice were unresponsive to administration of a CSF1-Fc fusion protein, which expanded tissue macrophage populations in controls. In the brain, microglial cell numbers and dendritic arborisation were reduced in Csf1rE631K/+ mice, as in patients with ALSP. The microglial phenotype is the opposite of microgliosis observed in Csf1r+/- mice. However, we found no evidence of brain pathology or impacts on motor function in aged Csf1rE631K/+ mice. We conclude that heterozygous disease-associated CSF1R mutations compromise CSF1R signalling. We speculate that leukoencephalopathy associated with dominant human CSF1R mutations requires an environmental trigger and/or epistatic interaction with common neurodegenerative disease-associated alleles.
Collapse
Affiliation(s)
- Jennifer Stables
- Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, Qld 4102, Australia
| | - Emma K Green
- Centre for Inflammation Research and Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Anuj Sehgal
- Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, Qld 4102, Australia
| | - Omkar L Patkar
- Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, Qld 4102, Australia
| | - Sahar Keshvari
- Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, Qld 4102, Australia
| | - Isis Taylor
- Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, Qld 4102, Australia
| | - Maisie E Ashcroft
- Centre for Inflammation Research and Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Kathleen Grabert
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Evi Wollscheid-Lengeling
- Luxembourg Centre for Systems Biomedicine, Université du Luxembourg, Belvaux, L-4401, Luxembourg
| | - Stefan Szymkowiak
- UK Dementia Research Institute, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Barry W McColl
- UK Dementia Research Institute, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Antony Adamson
- Genome Editing Unit, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Neil E Humphreys
- Genome Editing Unit, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Werner Mueller
- Genome Editing Unit, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Hana Starobova
- Institute for Molecular Biosciences & School of Pharmacy, University of Queensland, Brisbane, Qld 4072, Australia
| | - Irina Vetter
- Institute for Molecular Biosciences & School of Pharmacy, University of Queensland, Brisbane, Qld 4072, Australia
| | | | | | - Kim M Summers
- Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, Qld 4102, Australia
| | - Katharine M Irvine
- Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, Qld 4102, Australia
| | - Clare Pridans
- Centre for Inflammation Research and Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - David A Hume
- Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, Qld 4102, Australia
| |
Collapse
|
19
|
Uyar O, Dominguez JM, Bordeleau M, Lapeyre L, Ibáñez FG, Vallières L, Tremblay ME, Corbeil J, Boivin G. Single-cell transcriptomics of the ventral posterolateral nucleus-enriched thalamic regions from HSV-1-infected mice reveal a novel microglia/microglia-like transcriptional response. J Neuroinflammation 2022; 19:81. [PMID: 35387656 PMCID: PMC8985399 DOI: 10.1186/s12974-022-02437-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/13/2022] [Indexed: 12/17/2022] Open
Abstract
Background Microglia participate in the immune response upon central nervous system (CNS) infections. However, the role of these cells during herpes simplex encephalitis (HSE) has not been fully characterized. We sought to identify different microglia/microglia-like cells and describe the potential mechanisms and signaling pathways involved during HSE. Methods The transcriptional response of CD11b+ immune cells, including microglia/microglia-like cells, was investigated using single-cell RNA sequencing (scRNA-seq) on cells isolated from the ventral posterolateral nucleus (VPL)-enriched thalamic regions of C57BL/6 N mice intranasally infected with herpes simplex virus-1 (HSV-1) (6 × 105 PFUs/20 µl). We further performed scanning electronic microscopy (SEM) analysis in VPL regions on day 6 post-infection (p.i.) to provide insight into microglial functions. Results We describe a novel microglia-like transcriptional response associated with a rare cell population (7% of all analyzed cells), named “in transition” microglia/microglia-like cells in HSE. This new microglia-like transcriptional signature, found in the highly infected thalamic regions, was enriched in specific genes (Retnlg, Cxcr2, Il1f9) usually associated with neutrophils. Pathway analysis of this cell-type transcriptome showed increased NLRP3-inflammasome-mediated interleukin IL-1β production, promoting a pro-inflammatory response. These cells' increased expression of viral transcripts suggests that the distinct “in transition” transcriptome corresponds to the intrinsic antiviral immune signaling of HSV-1-infected microglia/microglia-like cells in the thalamus. In accordance with this phenotype, we observed several TMEM119+/IBA-I+ microglia/microglia-like cells immunostained for HSV-1 in highly infected regions. Conclusions A new microglia/microglia-like state may potentially shed light on how microglia could react to HSV-1 infection. Our observations suggest that infected microglia/microglia-like cells contribute to an exacerbated CNS inflammation. Further characterization of this transitory state of the microglia/microglia-like cell transcriptome may allow the development of novel immunomodulatory approaches to improve HSE outcomes by regulating the microglial immune response. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02437-7.
Collapse
Affiliation(s)
- Olus Uyar
- Research Center in Infectious Diseases, CHU de Québec-Laval University Research Center and Department of Pediatrics and Microbiology, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Juan Manuel Dominguez
- Research Center in Infectious Diseases, CHU de Québec-Laval University Research Center and Department of Molecular Medicine and Big Data Research Centre, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Maude Bordeleau
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.,Neurosciences Unit, CHU de Québec-Laval University Research Center, Quebec City, QC, Canada
| | - Lina Lapeyre
- Research Center in Infectious Diseases, CHU de Québec-Laval University Research Center and Department of Pediatrics and Microbiology, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Fernando González Ibáñez
- Neurosciences Unit, CHU de Québec-Laval University Research Center, Quebec City, QC, Canada.,Department of Molecular Medicine, Laval University, Quebec City, QC, Canada.,Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Luc Vallières
- Neurosciences Unit, CHU de Québec-Laval University Research Center, Quebec City, QC, Canada.,Department of Molecular Medicine, Laval University, Quebec City, QC, Canada
| | - Marie-Eve Tremblay
- Neurosciences Unit, CHU de Québec-Laval University Research Center, Quebec City, QC, Canada.,Department of Molecular Medicine, Laval University, Quebec City, QC, Canada.,Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.,Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada.,Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
| | - Jacques Corbeil
- Research Center in Infectious Diseases, CHU de Québec-Laval University Research Center and Department of Molecular Medicine and Big Data Research Centre, Faculty of Medicine, Laval University, Quebec City, QC, Canada.,Department of Molecular Medicine, Laval University, Quebec City, QC, Canada
| | - Guy Boivin
- Research Center in Infectious Diseases, CHU de Québec-Laval University Research Center and Department of Pediatrics and Microbiology, Faculty of Medicine, Laval University, Quebec City, QC, Canada.
| |
Collapse
|
20
|
Han J, Chitu V, Stanley ER, Wszolek ZK, Karrenbauer VD, Harris RA. Inhibition of colony stimulating factor-1 receptor (CSF-1R) as a potential therapeutic strategy for neurodegenerative diseases: opportunities and challenges. Cell Mol Life Sci 2022; 79:219. [PMID: 35366105 PMCID: PMC8976111 DOI: 10.1007/s00018-022-04225-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/06/2022] [Accepted: 02/26/2022] [Indexed: 12/12/2022]
Abstract
Microglia are specialized dynamic immune cells in the central nervous system (CNS) that plays a crucial role in brain homeostasis and in disease states. Persistent neuroinflammation is considered a hallmark of many neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson's disease (PD), Huntington’s disease (HD), amyotrophic lateral sclerosis (ALS) and primary progressive multiple sclerosis (MS). Colony stimulating factor 1-receptor (CSF-1R) is predominantly expressed on microglia and its expression is significantly increased in neurodegenerative diseases. Cumulative findings have indicated that CSF-1R inhibitors can have beneficial effects in preclinical neurodegenerative disease models. Research using CSF-1R inhibitors has now been extended into non-human primates and humans. This review article summarizes the most recent advances using CSF-1R inhibitors in different neurodegenerative conditions including AD, PD, HD, ALS and MS. Potential challenges for translating these findings into clinical practice are presented.
Collapse
Affiliation(s)
- Jinming Han
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Violeta Chitu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| | - E. Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| | | | - Virginija Danylaité Karrenbauer
- Department of Clinical Neuroscience, Center for Molecular Medicine L8:04, Karolinska Institutet, Karolinska University Hospital, 171 76 Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Robert A. Harris
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| |
Collapse
|
21
|
Small Noncoding RNA (sncRNA1) within the Latency-Associated Transcript Modulates Herpes Simplex Virus 1 Virulence and the Host Immune Response during Acute but Not Latent Infection. J Virol 2022; 96:e0005422. [DOI: 10.1128/jvi.00054-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
HSV-1 latency-associated transcript (LAT) plays a major role in establishing latency and reactivation; however, the mechanism by which LAT controls these processes is largely unknown. In this study, we sought to establish the role of the small noncoding RNA1 (sncRNA1) encoded within LAT during HSV-1 ocular infection. Our results suggest that sncRNA1 has a protective role during acute ocular infection by modulating the innate immune response to infection.
Collapse
|
22
|
O'Brien CA, Bennett FC, Bennett ML. Microglia in antiviral immunity of the brain and spinal cord. Semin Immunol 2022; 60:101650. [PMID: 36099864 PMCID: PMC9934594 DOI: 10.1016/j.smim.2022.101650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/17/2022] [Accepted: 08/30/2022] [Indexed: 01/15/2023]
Abstract
Viral infections of the central nervous system (CNS) are a significant cause of neurological impairment and mortality worldwide. As tissue resident macrophages, microglia are critical initial responders to CNS viral infection. Microglia seem to coordinate brain-wide antiviral responses of both brain resident cells and infiltrating immune cells. This review discusses how microglia may promote this antiviral response at a molecular level, from potential mechanisms of virus recognition to downstream cytokine responses and interaction with antiviral T cells. Recent advancements in genetic tools to specifically target microglia in vivo promise to further our understanding about the precise mechanistic role of microglia in CNS infection.
Collapse
Affiliation(s)
- Carleigh A O'Brien
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States.
| | - F Chris Bennett
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States; Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Mariko L Bennett
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States; Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| |
Collapse
|
23
|
Microglia activate early anti-viral responses upon HSV-1 entry into the brain to counteract development of encephalitis-like disease in mice. J Virol 2022; 96:e0131121. [PMID: 35045263 DOI: 10.1128/jvi.01311-21] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Spread of herpes simplex virus 1 (HSV1) from the periphery to the central nervous system (CNS) can lead to extensive infection and pathological inflammation in the brain, causing herpes simplex encephalitis (HSE). It has been shown that microglia, the CNS-resident macrophages, are involved in early sensing of HSV1 and an induction of antiviral responses. In addition, infiltration of peripheral immune cells may contribute to control of viral infection. In this study, we tested the effect of microglia depletion in a mouse model of HSE. Increased viral titers and increased disease severity were observed in microglia-depleted mice. The effect of microglia depletion was more pronounced in wild-type than in cGas-/- mice, revealing that this immune sensor contributes to the antiviral activity of microglia. Importantly, microglia depletion led to reduced production of type I interferon (IFN), pro-inflammatory cytokines and chemokines at early time points after viral entry into the CNS. In line with this, in vitro experiments on murine primary CNS cells demonstrated microglial presence to be essential for IFN RNA induction, and control of HSV1 replication. However, the effect of microglia depletion on expression of IFNs, and inflammatory cytokines was restricted to early time point of HSV1 entry into the CNS. There was no major alteration of infiltration of CD45-positive cells in microglia-depleted mice. Collectively, our data demonstrate a key role for microglia in controlling HSV1 replication early after viral entry into the CNS and highlight the importance of a prompt antiviral innate response to reduce the risk of HSE development. Importance One of the most devastating and acute neurological conditions is encephalitis, i.e. inflammation of brain tissue. Herpes simplex virus 1 (HSV1) is a highly prevalent pathogen in humans, and the most frequent cause of viral sporadic encephalitis, called herpes simplex encephalitis (HSE). HSV1 has the ability to infect peripheral neurons and reach the central nervous system (CNS) of humans, where it can be detected by brain resident cells and infiltrating immune cells, leading to protective and damaging immune responses. In this study, we investigated the effects of a depletion of microglia, the main brain-resident immune cell type. For this purpose, we used a mouse model of HSE. We found that viral levels increased and disease symptoms worsened in microglia-depleted mice. In addition, mice lacking a major sensor of viral DNA, cGAS, manifested more pronounced disease than wild-type mice, highlighting the importance of this immune sensor in the activity of microglia. Evidently, microglia depletion led to a reduced production of many known antiviral factors, most notably type I interferon (IFN). The importance of microglia in the early control of HSV1 spread and the generation of antiviral responses is further demonstrated by experiments on murine mixed glial cell cultures. Interestingly, mice with microglia depletion exhibited an unaltered activation of antiviral responses and recruitment of immune cells from the periphery at later time points of infection, but this did not prevent the development of the disease. Overall, the data highlight the importance of a rapid activation of the host defense, with microglia playing a critical role in controlling HSV1 infection, which eventually prevents damage to neurons and brain tissue.
Collapse
|
24
|
Tsai MS, Wang LC, Tsai HY, Lin YJ, Wu HL, Tzeng SF, Hsu SM, Chen SH. Microglia Reduce Herpes Simplex Virus 1 Lethality of Mice with Decreased T Cell and Interferon Responses in Brains. Int J Mol Sci 2021; 22:ijms222212457. [PMID: 34830340 PMCID: PMC8624831 DOI: 10.3390/ijms222212457] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/09/2021] [Accepted: 11/16/2021] [Indexed: 12/11/2022] Open
Abstract
Herpes simplex virus 1 (HSV-1) infects the majority of the human population and can induce encephalitis, which is the most common cause of sporadic, fatal encephalitis. An increase of microglia is detected in the brains of encephalitis patients. The issues regarding whether and how microglia protect the host and neurons from HSV-1 infection remain elusive. Using a murine infection model, we showed that HSV-1 infection on corneas increased the number of microglia to outnumber those of infiltrating leukocytes (macrophages, neutrophils, and T cells) and enhanced microglia activation in brains. HSV-1 antigens were detected in brain neurons, which were surrounded by microglia. Microglia depletion increased HSV-1 lethality of mice with elevated brain levels of viral loads, infected neurons, neuron loss, CD4 T cells, CD8 T cells, neutrophils, interferon (IFN)-β, and IFN-γ. In vitro studies demonstrated that microglia from infected mice reduced virus infectivity. Moreover, microglia induced IFN-β and the signaling pathway of signal transducer and activator of transcription (STAT) 1 to inhibit viral replication and damage of neurons. Our study reveals how microglia protect the host and neurons from HSV-1 infection.
Collapse
Affiliation(s)
- Meng-Shan Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; (M.-S.T.); (H.-L.W.)
| | - Li-Chiu Wang
- School of Medicine, I-Shou University, Kaohsiung 824, Taiwan;
| | - Hsien-Yang Tsai
- Department of Ophthalmology, Tzu Chi Hospital, Taichung 427, Taiwan;
| | - Yu-Jheng Lin
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan;
| | - Hua-Lin Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; (M.-S.T.); (H.-L.W.)
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Shun-Fen Tzeng
- Department of Life Sciences, College of Biological Science and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan;
| | - Sheng-Min Hsu
- Department of Ophthalmology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Correspondence: (S.-M.H.); (S.-H.C.)
| | - Shun-Hua Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; (M.-S.T.); (H.-L.W.)
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan;
- Correspondence: (S.-M.H.); (S.-H.C.)
| |
Collapse
|
25
|
Sehgal A, Irvine KM, Hume DA. Functions of macrophage colony-stimulating factor (CSF1) in development, homeostasis, and tissue repair. Semin Immunol 2021; 54:101509. [PMID: 34742624 DOI: 10.1016/j.smim.2021.101509] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 10/23/2021] [Indexed: 12/16/2022]
Abstract
Macrophage colony-stimulating factor (CSF1) is the primary growth factor required for the control of monocyte and macrophage differentiation, survival, proliferation and renewal. Although the cDNAs encoding multiple isoforms of human CSF1 were cloned in the 1980s, and recombinant proteins were available for testing in humans, CSF1 has not yet found substantial clinical application. Here we present an overview of CSF1 biology, including evolution, regulation and functions of cell surface and secreted isoforms. CSF1 is widely-expressed, primarily by cells of mesenchymal lineages, in all mouse tissues. Cell-specific deletion of a floxed Csf1 allele in mice indicates that local CSF1 production contributes to the maintenance of tissue-specific macrophage populations but is not saturating. CSF1 in the circulation is controlled primarily by receptor-mediated clearance by macrophages in liver and spleen. Administration of recombinant CSF1 to humans or animals leads to monocytosis and expansion of tissue macrophage populations and growth of the liver and spleen. In a wide variety of tissue injury models, CSF1 administration promotes monocyte infiltration, clearance of damaged cells and repair. We suggest that CSF1 has therapeutic potential in regenerative medicine.
Collapse
Affiliation(s)
- Anuj Sehgal
- Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Katharine M Irvine
- Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - David A Hume
- Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia.
| |
Collapse
|
26
|
Uyar O, Plante PL, Piret J, Venable MC, Carbonneau J, Corbeil J, Boivin G. A novel bioluminescent herpes simplex virus 1 for in vivo monitoring of herpes simplex encephalitis. Sci Rep 2021; 11:18688. [PMID: 34548521 PMCID: PMC8455621 DOI: 10.1038/s41598-021-98047-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 08/26/2021] [Indexed: 11/22/2022] Open
Abstract
Herpes simplex virus 1 (HSV-1) is responsible for herpes simplex virus encephalitis (HSE), associated with a 70% mortality rate in the absence of treatment. Despite intravenous treatment with acyclovir, mortality remains significant, highlighting the need for new anti-herpetic agents. Herein, we describe a novel neurovirulent recombinant HSV-1 (rHSV-1), expressing the fluorescent tdTomato and Gaussia luciferase (Gluc) enzyme, generated by the Clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9) (CRISPR-Cas9) system. The Gluc activity measured in the cell culture supernatant was correlated (P = 0.0001) with infectious particles, allowing in vitro monitoring of viral replication kinetics. A significant correlation was also found between brain viral titers and Gluc activity in plasma (R2 = 0.8510, P < 0.0001) collected from BALB/c mice infected intranasally with rHSV-1. Furthermore, evaluation of valacyclovir (VACV) treatment of HSE could also be performed by analyzing Gluc activity in mouse plasma samples. Finally, it was also possible to study rHSV-1 dissemination and additionally to estimate brain viral titers by in vivo imaging system (IVIS). The new rHSV-1 with reporter proteins is not only as a powerful tool for in vitro and in vivo antiviral screening, but can also be used for studying different aspects of HSE pathogenesis.
Collapse
Affiliation(s)
- Olus Uyar
- Research Center in Infectious Diseases, CHU de Québec-Laval University Research Center and Department of Pediatrics and Microbiology, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Pier-Luc Plante
- Research Center in Infectious Diseases, CHU de Québec-Laval University Research Center and Department of Molecular Medicine and Big Data Research Centre, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Jocelyne Piret
- Research Center in Infectious Diseases, CHU de Québec-Laval University Research Center and Department of Pediatrics and Microbiology, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Marie-Christine Venable
- Research Center in Infectious Diseases, CHU de Québec-Laval University Research Center and Department of Pediatrics and Microbiology, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Julie Carbonneau
- Research Center in Infectious Diseases, CHU de Québec-Laval University Research Center and Department of Pediatrics and Microbiology, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Jacques Corbeil
- Research Center in Infectious Diseases, CHU de Québec-Laval University Research Center and Department of Molecular Medicine and Big Data Research Centre, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Guy Boivin
- Research Center in Infectious Diseases, CHU de Québec-Laval University Research Center and Department of Pediatrics and Microbiology, Faculty of Medicine, Laval University, Quebec City, QC, Canada.
| |
Collapse
|
27
|
Krzyzowska M, Kowalczyk A, Skulska K, Thörn K, Eriksson K. Fas/FasL Contributes to HSV-1 Brain Infection and Neuroinflammation. Front Immunol 2021; 12:714821. [PMID: 34526992 PMCID: PMC8437342 DOI: 10.3389/fimmu.2021.714821] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/16/2021] [Indexed: 12/14/2022] Open
Abstract
The Fas/FasL pathway plays a key role in immune homeostasis and immune surveillance. In the central nervous system (CNS) Fas/FasL is involved in axonal outgrowth and adult neurogenesis. However, little is known about the role of the Fas/FasL pathway in herpes encephalitis. In this study, we used a neuropathogenic clinical strain of herpes simplex virus type 1 (HSV-1) to explore infection-induced inflammation and immune responses in the mouse brain and the role of Fas/FasL in antiviral CNS immunity. HSV-1 CNS infection induced the infiltration of Fas- FasL-bearing monocytes and T cells in the brain and also to an up-regulation of Fas and FasL expression on resident astrocytes and microglia within infected sites. Upon infection, Fas- and FasL-deficient mice (lpr and gld) were partially protected from encephalitis with a decreased morbidity and mortality compared to WT mice. Fas/FasL deficiency promoted cell-mediated immunity within the CNS. Fas receptor stimulation abrogated HSV-1 induced activation and inflammatory reactions in microglia from WT mice, while lack of Fas or FasL led to a more pronounced activation of monocytes and microglia and also to an enhanced differentiation of these cells into a pro-inflammatory M1 phenotype. Furthermore, the specific immune system was more efficient in Fas- and FasL-deficient mice with significantly higher numbers of infiltrating HSV-1-specific cytotoxic T cells in the brain. Our data indicate that the Fas/FasL pathway leads to excessive neuroinflammation during HSV-1 infection, which is associated with a diminished anti-viral response and an excessive neuroinflammation.
Collapse
Affiliation(s)
- Malgorzata Krzyzowska
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Virology and Cell Biology, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland.,Laboratory of Nanobiology and Biomaterials, Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| | - Andrzej Kowalczyk
- Department of Virology and Cell Biology, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Katarzyna Skulska
- Department of Virology and Cell Biology, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Karolina Thörn
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kristina Eriksson
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
28
|
The Influence of Virus Infection on Microglia and Accelerated Brain Aging. Cells 2021; 10:cells10071836. [PMID: 34360004 PMCID: PMC8303900 DOI: 10.3390/cells10071836] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
Microglia are the resident immune cells of the central nervous system contributing substantially to health and disease. There is increasing evidence that inflammatory microglia may induce or accelerate brain aging, by interfering with physiological repair and remodeling processes. Many viral infections affect the brain and interfere with microglia functions, including human immune deficiency virus, flaviviruses, SARS-CoV-2, influenza, and human herpes viruses. Especially chronic viral infections causing low-grade neuroinflammation may contribute to brain aging. This review elucidates the potential role of various neurotropic viruses in microglia-driven neurocognitive deficiencies and possibly accelerated brain aging.
Collapse
|