1
|
Hazra B, Das Sarma J. The CD40/CD40 ligand dyad and its downstream effector molecule ISG54 in relating acute neuroinflammation with persistent, progressive demyelination. IUBMB Life 2024; 76:313-331. [PMID: 38116887 DOI: 10.1002/iub.2798] [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: 02/08/2023] [Accepted: 11/14/2023] [Indexed: 12/21/2023]
Abstract
Although Multiple Sclerosis (MS) is primarily thought to be an autoimmune condition, its possible viral etiology must be taken into consideration. When mice are administered neurotropic viruses like mouse hepatitis virus MHV-A59, a murine coronavirus, or its isogenic recombinant strain RSA59, neuroinflammation along with demyelination are observed, which are some of the significant manifestations of MS. MHV-A59/RSA59 induced neuroinflammation is one of the best-studied experimental animal models to understand the viral-induced demyelination concurrent with axonal loss. In this experimental animal model, one of the major immune checkpoint regulators is the CD40-CD40L dyad, which helps in mediating both acute-innate, innate-adaptive, and chronic-adaptive immune responses. Hence, they are essential in reducing acute neuroinflammation and chronic progressive adaptive demyelination. While CD40 is expressed on antigen-presenting cells and endothelial cells, CD40L is expressed primarily on activated T cells and during severe inflammation on NK cells and mast cells. Experimental evidences revealed that genetic deficiency of both these proteins can lead to deleterious effects in an individual. On the other hand, interferon-stimulated genes (ISGs) possess potent antiviral properties and directly or indirectly alter acute neuroinflammation. In this review, we will discuss the role of an ISG, ISG54, and its tetratricopeptide repeat protein Ifit2; the genetic and experimental studies on the role of CD40 and CD40L in a virus-induced neuroinflammatory demyelination model.
Collapse
Affiliation(s)
- Bishal Hazra
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
2
|
Guérin A, Moncada-Vélez M, Jackson K, Ogishi M, Rosain J, Mancini M, Langlais D, Nunez A, Webster S, Goyette J, Khan T, Marr N, Avery DT, Rao G, Waterboer T, Michels B, Neves E, Iracema Morais C, London J, Mestrallet S, Quartier dit Maire P, Neven B, Rapaport F, Seeleuthner Y, Lev A, Simon AJ, Montoya J, Barel O, Gómez-Rodríguez J, Orrego JC, L’Honneur AS, Soudée C, Rojas J, Velez AC, Sereti I, Terrier B, Marin N, García LF, Abel L, Boisson-Dupuis S, Reis J, Marinho A, Lisco A, Faria E, Goodnow CC, Vasconcelos J, Béziat V, Ma CS, Somech R, Casanova JL, Bustamante J, Franco JL, Tangye SG. Helper T cell immunity in humans with inherited CD4 deficiency. J Exp Med 2024; 221:e20231044. [PMID: 38557723 PMCID: PMC10983808 DOI: 10.1084/jem.20231044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 01/04/2024] [Accepted: 01/31/2024] [Indexed: 04/04/2024] Open
Abstract
CD4+ T cells are vital for host defense and immune regulation. However, the fundamental role of CD4 itself remains enigmatic. We report seven patients aged 5-61 years from five families of four ancestries with autosomal recessive CD4 deficiency and a range of infections, including recalcitrant warts and Whipple's disease. All patients are homozygous for rare deleterious CD4 variants impacting expression of the canonical CD4 isoform. A shorter expressed isoform that interacts with LCK, but not HLA class II, is affected by only one variant. All patients lack CD4+ T cells and have increased numbers of TCRαβ+CD4-CD8- T cells, which phenotypically and transcriptionally resemble conventional Th cells. Finally, patient CD4-CD8- αβ T cells exhibit intact responses to HLA class II-restricted antigens and promote B cell differentiation in vitro. Thus, compensatory development of Th cells enables patients with inherited CD4 deficiency to acquire effective cellular and humoral immunity against an unexpectedly large range of pathogens. Nevertheless, CD4 is indispensable for protective immunity against at least human papillomaviruses and Trophyrema whipplei.
Collapse
Affiliation(s)
- Antoine Guérin
- Garvan Institute of Medical Research, Darlinghurst, Australia
- Faculty of Medicine and Health, School of Clinical Medicine, University of New South Wales Sydney, Sydney, Australia
| | - Marcela Moncada-Vélez
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Primary Immunodeficiencies Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellin, Colombia
| | | | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Jérémie Rosain
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - Mathieu Mancini
- Department of Human Genetics, McGill University, Montreal, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
- Dahdaleh Institute of Genomic Medicine, McGill Research Centre on Complex Traits, McGill University, Montreal, Canada
| | - David Langlais
- Department of Human Genetics, McGill University, Montreal, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
- Dahdaleh Institute of Genomic Medicine, McGill Research Centre on Complex Traits, McGill University, Montreal, Canada
| | - Andrea Nunez
- Department of Molecular Medicine, School of Biomedical Sciences, University of New South Wales, Sydney, Australia
| | - Samantha Webster
- Department of Molecular Medicine, School of Biomedical Sciences, University of New South Wales, Sydney, Australia
| | - Jesse Goyette
- Department of Molecular Medicine, School of Biomedical Sciences, University of New South Wales, Sydney, Australia
| | - Taushif Khan
- Department of Human Immunology, Sidra Medicine, Doha, Qatar
- The Jackson Laboratory, Farmington, CT, USA
| | - Nico Marr
- Department of Human Immunology, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Danielle T. Avery
- Garvan Institute of Medical Research, Darlinghurst, Australia
- Faculty of Medicine and Health, School of Clinical Medicine, University of New South Wales Sydney, Sydney, Australia
| | - Geetha Rao
- Garvan Institute of Medical Research, Darlinghurst, Australia
- Faculty of Medicine and Health, School of Clinical Medicine, University of New South Wales Sydney, Sydney, Australia
| | - Tim Waterboer
- Division of Infections and Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Birgitta Michels
- Division of Infections and Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Esmeralda Neves
- Immunology Department—Pathology, University Hospital Center of Porto, Porto, Portugal
- Unit for Multidisciplinary Research in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Cátia Iracema Morais
- Immunology Department—Pathology, University Hospital Center of Porto, Porto, Portugal
- Unit for Multidisciplinary Research in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Jonathan London
- Service of Internal Medicine, Diaconesse-Croix Saint Simon Hospital, Paris, France
| | - Stéphanie Mestrallet
- Department of Internal Medicine and Infectious Diseases, Manchester Hospital, Charleville-Mézières, France
| | - Pierre Quartier dit Maire
- Pediatric Immunology-Hematology and Rheumatology Unit, Necker Hospital for Sick Children, Paris, France
| | - Bénédicte Neven
- Pediatric Immunology-Hematology and Rheumatology Unit, Necker Hospital for Sick Children, Paris, France
| | - Franck Rapaport
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Yoann Seeleuthner
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Atar Lev
- Department of Pediatrics and Immunology Service, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Aviv School of Medicine, Tel Aviv, Israel
| | - Amos J. Simon
- Department of Pediatrics and Immunology Service, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Aviv School of Medicine, Tel Aviv, Israel
| | - Jorge Montoya
- San Vicente de Paul University Hospital, Medellin, Colombia
| | - Ortal Barel
- The Genomic Unit, Sheba Cancer Research Center, Sheba Medical Center, Ramat Gan, Israel
| | - Julio Gómez-Rodríguez
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Julio C. Orrego
- Primary Immunodeficiencies Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellin, Colombia
| | - Anne-Sophie L’Honneur
- Department of Virology, Paris Cité University and Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Camille Soudée
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Jessica Rojas
- Primary Immunodeficiencies Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellin, Colombia
| | - Alejandra C. Velez
- Primary Immunodeficiencies Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellin, Colombia
| | - Irini Sereti
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin Terrier
- Department of Internal Medicine, Cochin Hospital, Assistance Publique–Hôpitaux de Paris, Paris Cité University, Paris, France
| | - Nancy Marin
- Cellular Immunology and Immunogenetics Group, University of Antioquia UdeA, Medellin, Colombia
| | - Luis F. García
- Cellular Immunology and Immunogenetics Group, University of Antioquia UdeA, Medellin, Colombia
| | - Laurent Abel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Stéphanie Boisson-Dupuis
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Joel Reis
- Dermatology Service, University Hospital Center of Porto, Porto, Portugal
| | - Antonio Marinho
- School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
- Department of Clinical Immunology, University Hospital Center of Porto, Porto, Portugal
| | - Andrea Lisco
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Emilia Faria
- Allergy and Clinical Immunology Department, University Hospital Center of Coimbra, Coimbra, Portugal
| | - Christopher C. Goodnow
- Garvan Institute of Medical Research, Darlinghurst, Australia
- Faculty of Medicine and Health, School of Clinical Medicine, University of New South Wales Sydney, Sydney, Australia
| | - Julia Vasconcelos
- Immunology Department—Pathology, University Hospital Center of Porto, Porto, Portugal
- Unit for Multidisciplinary Research in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Vivien Béziat
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Cindy S. Ma
- Garvan Institute of Medical Research, Darlinghurst, Australia
- Faculty of Medicine and Health, School of Clinical Medicine, University of New South Wales Sydney, Sydney, Australia
| | - Raz Somech
- Department of Pediatrics and Immunology Service, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Aviv School of Medicine, Tel Aviv, Israel
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Howard Hughes Medical Institute, New York, NY, USA
- Department of Pediatrics, Necker Hospital for Sick Children, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - Jacinta Bustamante
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - Jose Luis Franco
- Primary Immunodeficiencies Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellin, Colombia
| | - Stuart G. Tangye
- Garvan Institute of Medical Research, Darlinghurst, Australia
- Faculty of Medicine and Health, School of Clinical Medicine, University of New South Wales Sydney, Sydney, Australia
| |
Collapse
|
3
|
Sharma M, Chakravarty D, Hussain A, Zalavadia A, Burrows A, Rayman P, Sharma N, Kenyon LC, Bergmann C, Sen GC, Das Sarma J. Ifit2 restricts murine coronavirus spread to the spinal cord white matter and its associated myelin pathology. J Virol 2023; 97:e0074923. [PMID: 37504572 PMCID: PMC10506381 DOI: 10.1128/jvi.00749-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 06/11/2023] [Indexed: 07/29/2023] Open
Abstract
Interferon-induced protein with tetratricopeptide repeats 2, Ifit2, is critical in restricting neurotropic murine-β-coronavirus, RSA59 infection. RSA59 intracranial injection of Ifit2-deficient (-/-) compared to wild-type (WT) mice results in impaired acute microglial activation, reduced CX3CR1 expression, limited migration of peripheral lymphocytes into the brain, and impaired virus control followed by severe morbidity and mortality. While the protective role of Ifit2 is established for acute viral encephalitis, less is known about its influence during the chronic demyelinating phase of RSA59 infection. To understand this, RSA59 infected Ifit2-/- and Ifit2+/+ (WT) were observed for neuropathological outcomes at day 5 (acute phase) and 30 post-infection (chronic phase). Our study demonstrates that Ifit2 deficiency causes extensive RSA59 spread throughout the spinal cord gray and white matter, associated with impaired CD4+ T and CD8+ T cell infiltration. Further, the cervical lymph nodes of RSA59 infected Ifit2-/- mice showed reduced activation of CD4+ T cells and impaired IFNγ expression during acute encephalomyelitis. Interestingly, BBB integrity was better preserved in Ifit2-/- mice, as evidenced by tight junction protein Claudin-5 and adapter protein ZO-1 expression surrounding the meninges and blood vessels and decreased Texas red dye uptake, which may be responsible for reduced leukocyte infiltration. In contrast to sparse myelin loss in WT mice, the chronic disease phase in Ifit2-/- mice was associated with severe demyelination and persistent viral load, even at low inoculation doses. Overall, our study highlights that Ifit2 provides antiviral functions by promoting acute neuroinflammation and thereby aiding virus control and limiting severe chronic demyelination. IMPORTANCE Interferons execute their function by inducing specific genes collectively termed as interferon-stimulated genes (ISGs), among which interferon-induced protein with tetratricopeptide repeats 2, Ifit2, is known for restricting neurotropic viral replication and spread. However, little is known about its role in viral spread to the spinal cord and its associated myelin pathology. Toward this, our study using a neurotropic murine β-coronavirus and Ifit2-deficient mice demonstrates that Ifit2 deficiency causes extensive viral spread throughout the gray and white matter of the spinal cord accompanied by impaired microglial activation and T cell infiltration. Furthermore, infected Ifit2-deficient mice showed impaired activation of T cells in the cervical lymph node and relatively intact blood-brain barrier integrity. Overall, Ifit2 plays a crucial role in mounting host immunity against neurotropic murine coronavirus in the acute phase while preventing mice from developing viral-induced severe chronic neuroinflammatory demyelination, the characteristic feature of human neurological disease multiple sclerosis (MS).
Collapse
Affiliation(s)
- Madhav Sharma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Debanjana Chakravarty
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Afaq Hussain
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Ajay Zalavadia
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Amy Burrows
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Patricia Rayman
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Nikhil Sharma
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Lawrence C Kenyon
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | - Ganes C. Sen
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| |
Collapse
|
4
|
Kamble M, Saadi F, Kumar S, Saha B, Das Sarma J. Inducible nitric oxide synthase deficiency promotes murine-β-coronavirus induced demyelination. Virol J 2023; 20:51. [PMID: 36966345 PMCID: PMC10039690 DOI: 10.1186/s12985-023-02006-1] [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: 11/11/2022] [Accepted: 03/06/2023] [Indexed: 03/27/2023] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is characterized by neuroinflammation and demyelination orchestrated by activated neuroglial cells, CNS infiltrating leukocytes, and their reciprocal interactions through inflammatory signals. An inflammatory stimulus triggers inducible nitric oxide synthase (NOS2), a pro-inflammatory marker of microglia/macrophages (MG/Mφ) to catalyze sustained nitric oxide production. NOS2 during neuroinflammation, has been associated with MS disease pathology; however, studies dissecting its role in demyelination are limited. We studied the role of NOS2 in a recombinant β-coronavirus-MHV-RSA59 induced neuroinflammation, an experimental animal model mimicking the pathological hallmarks of MS: neuroinflammatory demyelination and axonal degeneration. OBJECTIVE Understanding the role of NOS2 in murine-β-coronavirus-MHV-RSA59 demyelination. METHODS Brain and spinal cords from mock and RSA59 infected 4-5-week-old MHV-free C57BL/6 mice (WT) and NOS2-/- mice were harvested at different disease phases post infection (p.i.) (day 5/6-acute, day 9/10-acute-adaptive and day 30-chronic phase) and compared for pathological outcomes. RESULTS NOS2 was upregulated at the acute phase of RSA59-induced disease in WT mice and its deficiency resulted in severe disease and reduced survival at the acute-adaptive transition phase. Low survival in NOS2-/- mice was attributed to (i) high neuroinflammation resulting from increased accumulation of macrophages and neutrophils and (ii) Iba1 + phagocytic MG/Mφ mediated-early demyelination as observed at this phase. The phagocytic phenotype of CNS MG/Mφ was confirmed by significantly higher mRNA transcripts of phagocyte markers-CD206, TREM2, and Arg1 and double immunolabelling of Iba1 with MBP and PLP. Further, NOS2 deficiency led to exacerbated demyelination at the chronic phase as well. CONCLUSION Taken together the results imply that the immune system failed to control the disease progression in the absence of NOS2. Thus, our observations highlight a protective role of NOS2 in murine-β-coronavirus induced demyelination.
Collapse
Affiliation(s)
- Mithila Kamble
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Fareeha Saadi
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Saurav Kumar
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Bhaskar Saha
- National Centre for Cell Science, Ganeshkhind, Pune, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India.
- Department of Ophthalmology, University of Pennsylvania Scheie Eye Institute, Philadelphia, PA, 19104, USA.
| |
Collapse
|
5
|
Liang W, Wang X, Xie N, Yan H, Ma H, Liu M, Kong W, Zhu Z, Bai W, Xiang H. Short-term associations of PM 2.5 and PM 2.5 constituents with immune biomarkers: A panel study in people living with HIV/AIDS. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 317:120743. [PMID: 36442818 DOI: 10.1016/j.envpol.2022.120743] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/07/2022] [Accepted: 11/24/2022] [Indexed: 06/16/2023]
Abstract
Studies on associations of fine particulate matter (PM2.5) with immunity in people living with HIV/AIDS (PLWHA) were absent. We aimed to explore whether changes of immune biomarkers were associated with short-term exposure to PM2.5 in PLWHA. Based on a panel study in Wuhan, we selected 163 PLWHA as participants with up to 4 repeated visits from March 2020 to January 2021. Immune biomarkers, including CD4+T cell count, CD8+T cell count, HIV viral load (VL) and CD4+T/CD8+T ratio were tested for all participants at each visit. Residential exposures of PM2.5 and PM2.5 constituents for each participant were assessed using spatial-temporal models. Linear mixed-effect models and general linear mixed models were applied to evaluate the associations between PM2.5 and immune biomarkers. To estimate the combined effect of PM2.5 constituents, weighted quantile sum regression and Bayesian kernel machine regression were employed. Each 10 μg/m3 increase of 7-day average PM2.5 concentrations was associated with an 8.75 cells/mm3 (95%CI: -15.55, -1.98) decrease in CD4+T cell count and a 92% (OR: 1.92, 95%CI: 1.43, 2.58) increased odds ratio of detectable HIV VL. However, the odds ratio of inverted CD4+T/CD8+T was only positively associated with PM2.5 concentrations at lag2 day (OR:1.27, 95%CI:1.02, 1.57). CD4+T may be a potential mediator between PM2.5 and detectable HIV VL with 3.83% mediated proportion. Besides, the combined effect of PM2.5 chemical constituents indicated that NO3- and SO42- were the main constituents in reducing CD4+T cell count and increasing odds ratio of detectable HIV VL. Our finding revealed that short-term exposure to PM2.5 was negatively associated with CD4+T cell count but positively related to the odds ratio of detectable HIV VL in PLWHA. This research may provide new evidence in associations between PM2.5 and immune biomarkers as well as improving prognosis of PLWHA.
Collapse
Affiliation(s)
- Wei Liang
- Department of Global Health, School of Public Health, Wuhan University, 115# Donghu Road, Wuhan, 430071, China; Global Health Institute, School of Public Health, Wuhan University, 115# Donghu Road, Wuhan, 430071, China
| | - Xia Wang
- Wuhan Center for Disease Control and Prevention, 288# Machang Road, Wuhan, 430024, China
| | - Nianhua Xie
- Wuhan Center for Disease Control and Prevention, 288# Machang Road, Wuhan, 430024, China
| | - Han Yan
- Wuhan Center for Disease Control and Prevention, 288# Machang Road, Wuhan, 430024, China
| | - Hongfei Ma
- Wuhan Center for Disease Control and Prevention, 288# Machang Road, Wuhan, 430024, China
| | - Manqing Liu
- Wuhan Center for Disease Control and Prevention, 288# Machang Road, Wuhan, 430024, China
| | - Wenhua Kong
- Wuhan Center for Disease Control and Prevention, 288# Machang Road, Wuhan, 430024, China
| | - Zerong Zhu
- Wuhan Center for Disease Control and Prevention, 288# Machang Road, Wuhan, 430024, China
| | - Wenjuan Bai
- Wuhan Center for Disease Control and Prevention, 288# Machang Road, Wuhan, 430024, China
| | - Hao Xiang
- Department of Global Health, School of Public Health, Wuhan University, 115# Donghu Road, Wuhan, 430071, China; Global Health Institute, School of Public Health, Wuhan University, 115# Donghu Road, Wuhan, 430071, China.
| |
Collapse
|
6
|
Nakayama M, Kyuwa S. Basic reproduction numbers of three strains of mouse hepatitis viruses in mice. Microbiol Immunol 2022; 66:166-172. [PMID: 34984727 PMCID: PMC9306726 DOI: 10.1111/1348-0421.12961] [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: 10/08/2021] [Revised: 11/29/2021] [Accepted: 12/26/2021] [Indexed: 11/26/2022]
Abstract
Mouse hepatitis virus (MHV) is a murine coronavirus and one of the most important pathogens in laboratory mice. Although various strains of MHV have been isolated, they are generally excreted in the feces and transmitted oronasally via aerosols and contaminated bedding. In this study, we attempted to determine the basic reproduction numbers (R0) of three strains of MHV to improve our understanding of MHV infections in mice. Five‐week‐old female C57BL/6J mice were inoculated intranasally with either the Y, NuU, or JHM variant strain of MHV and housed with two naïve mice. After 4 weeks, the presence or absence of anti‐MHV antibody in the mice was determined by ELISA. We also examined the distribution of MHV in the organs of Y, NuU, or JHM variant‐infected mice. Our data suggest that the transmissibility of MHV is correlated with viral growth in the gastrointestinal tract of infected mice. To the best of our knowledge, this is the first report to address the basic reproduction numbers among pathogens in laboratory animals.
Collapse
Affiliation(s)
- Masataka Nakayama
- Laboratory of Biomedical Science, Department of Veterinary Medical Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Shigeru Kyuwa
- Laboratory of Biomedical Science, Department of Veterinary Medical Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
7
|
Sengupta S, Addya S, Biswas D, Banerjee P, Sarma JD. Matrix metalloproteinases and tissue inhibitors of metalloproteinases in murine β-coronavirus-induced neuroinflammation. Virology 2022; 566:122-135. [PMID: 34906793 PMCID: PMC8648396 DOI: 10.1016/j.virol.2021.11.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/16/2021] [Accepted: 11/26/2021] [Indexed: 01/17/2023]
Abstract
Mouse hepatitis virus (MHV; m-β-CoV) serves as a useful model for studying the cellular factors involved in neuroinflammation. To understand the role of matrix metalloproteinases (MMPs) in neuroinflammation, brain tissues from m-β-CoV-infected mice were harvested at different days post-infection (d.p.i) and investigated for Mmp expression by RT-qPCR. Mmp-2, -3, -8, -12 showed significant mRNA upregulation peaking with viral replication between 5 and 6 d.p.i. Elevated levels of MMP regulator TIMP-1 are suggestive of a TIMP-1 mediated host antiviral response. Biological network assessment suggested a direct involvement of MMP-3, -8, -14 in facilitating peripheral leukocyte infiltrations. Flow cytometry confirmed the increased presence of NK cells, CD4+ and CD8+ T cells, neutrophils, and MHCII expressing cells in the m-β-CoV infected mice brain. Our study revealed that m-β-CoV upregulated Park7, RelA, Nrf2, and Hmox1 transcripts involved in ROS production and antioxidant pathways, describing the possible nexus between oxidative pathways, MMPs, and TIMP in m-β-CoV-induced neuroinflammation.
Collapse
Affiliation(s)
- Sourodip Sengupta
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K), Mohanpur, India
| | - Sankar Addya
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA
| | - Diptomit Biswas
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K), Mohanpur, India
| | - Paromita Banerjee
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K), Mohanpur, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K), Mohanpur, India,Corresponding author
| |
Collapse
|
8
|
Saadi F, Chakravarty D, Kumar S, Kamble M, Saha B, Shindler KS, Das Sarma J. CD40L protects against mouse hepatitis virus-induced neuroinflammatory demyelination. PLoS Pathog 2021; 17:e1010059. [PMID: 34898656 PMCID: PMC8699621 DOI: 10.1371/journal.ppat.1010059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 12/23/2021] [Accepted: 10/23/2021] [Indexed: 11/19/2022] Open
Abstract
Neurotropic mouse hepatitis virus (MHV-A59/RSA59) infection in mice induces acute neuroinflammation due to direct neural cell dystrophy, which proceeds with demyelination with or without axonal loss, the pathological hallmarks of human neurological disease, Multiple sclerosis (MS). Recent studies in the RSA59-induced neuroinflammation model of MS showed a protective role of CNS-infiltrating CD4+ T cells compared to their pathogenic role in the autoimmune model. The current study further investigated the molecular nexus between CD4+ T cell-expressed CD40Ligand and microglia/macrophage-expressed CD40 using CD40L-/- mice. Results demonstrate CD40L expression in the CNS is modulated upon RSA59 infection. We show evidence that CD40L-/- mice are more susceptible to RSA59 induced disease due to reduced microglia/macrophage activation and significantly dampened effector CD4+ T recruitment to the CNS on day 10 p.i. Additionally, CD40L-/- mice exhibited severe demyelination mediated by phagocytic microglia/macrophages, axonal loss, and persistent poliomyelitis during chronic infection, indicating CD40-CD40L as host-protective against RSA59-induced demyelination. This suggests a novel target in designing prophylaxis for virus-induced demyelination and axonal degeneration, in contrast to immunosuppression which holds only for autoimmune mechanisms of inflammatory demyelination. MS is primarily considered an autoimmune CNS disease, but its potential viral etiology cannot be ignored. Myelin-specific CD40L+CD4+ T cells migration into the CNS and resultant neuroinflammation is considered pathogenic in autoimmune MS. In contrast, CD40L+CD4+ T infiltration into the MHV-induced inflamed CNS and their interaction with CD40+ microglia/macrophages are shown to be protective in our study. Considering differential etiology but comparable demyelination and axonal loss, immunosuppressive treatments may not necessarily ameliorate MS in all patients. MHV-induced demyelination in this study indicates that the interaction between CD40L on CD4+T cells and CD40 on microglia/macrophage plays an important protective role against MHV-induced chronic progressive demyelination.
Collapse
Affiliation(s)
- Fareeha Saadi
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Debanjana Chakravarty
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Saurav Kumar
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Mithila Kamble
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Bhaskar Saha
- National Centre for Cell Science, Ganeshkhind, Pune, India
| | - Kenneth S. Shindler
- Departments of Ophthalmology and
- Neurology University of Pennsylvania Scheie Eye Institute, Philadelphia, Pennsylvania, United States of America
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
- Departments of Ophthalmology and
- * E-mail:
| |
Collapse
|
9
|
Bakhtazad A, Garmabi B, Joghataei MT. Neurological manifestations of coronavirus infections, before and after COVID-19: a review of animal studies. J Neurovirol 2021; 27:864-884. [PMID: 34727365 PMCID: PMC8561685 DOI: 10.1007/s13365-021-01014-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 07/15/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This virus, which was first identified in December 2019 in China, has resulted in a yet ongoing viral pandemic. Coronaviridae could potentially cause several disorders in a wide range of hosts such as birds and mammals. Although infections caused by this family of viruses are predominantly limited to the respiratory tract, Betacoronaviruses are potentially able to invade the central nervous system (CNS) as well as many other organs, thereby inducing neurological damage ranging from mild to lethal in both animals and humans. Over the past two decades, three novel CoVs, SARS-CoV-1, MERS-CoV, and SARS-CoV-2, emerging from animal reservoirs have exhibited neurotropic properties causing severe and even fatal neurological diseases. The pathobiology of these neuroinvasive viruses has yet to be fully known. Both clinical features of the previous CoV epidemics (SARS-CoV-1 and MERS-CoV) and lessons from animal models used in studying neurotropic CoVs, especially SARS and MERS, constitute beneficial tools in comprehending the exact mechanisms of virus implantation and in illustrating pathogenesis and virus dissemination pathways in the CNS. Here, we review the animal research which assessed CNS infections with previous more studied neurotropic CoVs to demonstrate how experimental studies with appliable animal models can provide scientists with a roadmap in the CNS impacts of SARS-CoV-2. Indeed, animal studies can finally help us discover the underlying mechanisms of damage to the nervous system in COVID-19 patients and find novel therapeutic agents in order to reduce mortality and morbidity associated with neurological complications of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Atefeh Bakhtazad
- Cellular and Molecular Research Center (CMRC), Iran University of Medical Sciences, 1449614535 Tehran, Iran
| | - Behzad Garmabi
- School of Medicine, Shahroud University of Medical Sciences, Haft-Tir Sq, University Blv, 3614773947 Shahroud, Iran
| | - Mohammad Taghi Joghataei
- Cellular and Molecular Research Center (CMRC), Iran University of Medical Sciences, 1449614535 Tehran, Iran
| |
Collapse
|
10
|
Kundu S, Saadi F, Sengupta S, Antony GR, Raveendran VA, Kumar R, Kamble MA, Sarkar L, Burrows A, Pal D, Sen GC, Sarma JD. DJ-1-Nrf2 axis is activated upon murine β-coronavirus infection in the CNS. BRAIN DISORDERS 2021; 4:100021. [PMID: 34514445 PMCID: PMC8418700 DOI: 10.1016/j.dscb.2021.100021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 07/03/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022] Open
Abstract
Coronaviruses have emerged as alarming pathogens owing to their inherent ability of genetic variation and cross-species transmission. Coronavirus infection burdens the endoplasmic reticulum (ER.), causes reactive oxygen species production and induces host stress responses, including unfolded protein response (UPR) and antioxidant system. In this study, we have employed a neurotropic murine β-coronavirus (M-CoV) infection in the Central Nervous System (CNS) of experimental mice model to study the role of host stress responses mediated by interplay of DJ-1 and XBP1. DJ-1 is an antioxidant molecule with established functions in neurodegeneration. However, its regulation in virus-induced cellular stress response is less explored. Our study showed that M-CoV infection activated the glial cells and induced antioxidant and UPR genes during the acute stage when the viral titer peaks. As the virus particles decreased and acute neuroinflammation diminished at day ten p.i., a significant up-regulation in UPR responsive XBP1, antioxidant DJ-1, and downstream signaling molecules, including Nrf2, was recorded in the brain tissues. Additionally, preliminary in silico analysis of the binding between the DJ-1 promoter and a positively charged groove of XBP1 is also investigated, thus hinting at a mechanism behind the upregulation of DJ-1 during MHV-infection. The current study thus attempts to elucidate a novel interplay between the antioxidant system and UPR in the outcome of coronavirus infection.
Collapse
Affiliation(s)
- Soumya Kundu
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Fareeha Saadi
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Sourodip Sengupta
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Gisha Rose Antony
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Vineeth A Raveendran
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Rahul Kumar
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Mithila Ashok Kamble
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Lucky Sarkar
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Amy Burrows
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio, USA
| | - Debnath Pal
- Department of Computational and Data Sciences, Indian Institute of Science, Bangalore, India
| | - Ganes C Sen
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio, USA
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| |
Collapse
|
11
|
Saadi F, Pal D, Sarma JD. Spike Glycoprotein Is Central to Coronavirus Pathogenesis-Parallel Between m-CoV and SARS-CoV-2. Ann Neurosci 2021; 28:201-218. [PMID: 35341224 PMCID: PMC8948335 DOI: 10.1177/09727531211023755] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 03/24/2021] [Indexed: 01/04/2023] Open
Abstract
Coronaviruses (CoVs) are single-stranded, polyadenylated, enveloped RNA of positive polarity with a unique potential to alter host tropism. This has been exceptionally demonstrated by the emergence of deadly virus outbreaks of the past: Severe Acute Respiratory Syndrome (SARS-CoV) in 2003 and Middle East Respiratory Syndrome (MERS-CoV) in 2012. The 2019 outbreak by the new cross-species transmission of SARS-CoV-2 has put the world on alert. CoV infection is triggered by receptor recognition, membrane fusion, and successive viral entry mediated by the surface Spike (S) glycoprotein. S protein is one of the major antigenic determinants and the target for neutralizing antibodies. It is a valuable target in antiviral therapies because of its central role in cell-cell fusion, viral antigen spread, and host immune responses leading to immunopathogenesis. The receptor-binding domain of S protein has received greater attention as it initiates host attachment and contains major antigenic determinants. However, investigating the therapeutic potential of fusion peptide as a part of the fusion core complex assembled by the heptad repeats 1 and 2 (HR1 and HR2) is also warranted. Along with receptor attachment and entry, fusion mechanisms should also be explored for designing inhibitors as a therapeutic intervention. In this article, we review the S protein function and its role in mediating membrane fusion, spread, tropism, and its associated pathogenesis with notable therapeutic strategies focusing on results obtained from studies on a murine β-Coronavirus (m-CoV) and its associated disease process.
Collapse
Affiliation(s)
- Fareeha Saadi
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Kolkata, West Bengal, India
| | - Debnath Pal
- Department of Computational and Data Sciences, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Kolkata, West Bengal, India
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
12
|
Ryding M, Gamre M, Nissen MS, Nilsson AC, Okarmus J, Poulsen AAE, Meyer M, Blaabjerg M. Neurodegeneration Induced by Anti-IgLON5 Antibodies Studied in Induced Pluripotent Stem Cell-Derived Human Neurons. Cells 2021; 10:cells10040837. [PMID: 33917676 PMCID: PMC8068068 DOI: 10.3390/cells10040837] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 11/16/2022] Open
Abstract
Anti-IgLON5 disease is a progressive neurological disorder associated with autoantibodies against a neuronal cell adhesion molecule, IgLON5. In human postmortem brain tissue, the neurodegeneration and accumulation of hyperphosphorylated tau (p-tau) are found. Whether IgLON5 antibodies induce neurodegeneration or neurodegeneration provokes an immune response causing inflammation and antibody formation remains to be elucidated. We investigated the effects of anti-IgLON5 antibodies on human neurons. Human neural stem cells were differentiated for 14–48 days and exposed from Days 9 to 14 (short-term) or Days 13 to 48 (long-term) to either (i) IgG from a patient with confirmed anti-IgLON5 antibodies or (ii) IgG from healthy controls. The electrical activity of neurons was quantified using multielectrode array assays. Cultures were immunostained for β-tubulin III and p-tau and counterstained with 4′,6-Diamidine-2′-phenylindole dihydrochloride (DAPI). To study the impact on synapses, cultures were also immunostained for the synaptic proteins postsynaptic density protein 95 (PSD95) and synaptophysin. A lactate dehydrogenase release assay and nuclei morphology analysis were used to assess cell viability. Cultures exposed to anti-IgLON5 antibodies showed reduced neuronal spike rate and synaptic protein content, and the proportion of neurons with degenerative appearance including p-tau (T205)-positive neurons was higher when compared to cultures exposed to control IgG. In addition, cell death was increased in cultures exposed to anti-IgLON5 IgG for 21 days. In conclusion, pathological anti-IgLON5 antibodies induce neurodegenerative changes and cell death in human neurons. This supports the hypothesis that autoantibodies may induce the neurodegenerative changes found in patients with anti-IgLON5-mediated disease. Furthermore, this study highlights the potential use of stem cell-based in vitro models for investigations of antibody-mediated diseases. As anti-IgLON5 disease is heterogeneous, more studies with different IgLON5 antibody samples tested on human neurons are needed.
Collapse
Affiliation(s)
- Matias Ryding
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (M.R.); (M.G.); (J.O.); (M.M.)
- Department of Neurology, Odense University Hospital, 5000 Odense, Denmark; (M.S.N.); (A.A.E.P.)
| | - Mattias Gamre
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (M.R.); (M.G.); (J.O.); (M.M.)
| | - Mette S. Nissen
- Department of Neurology, Odense University Hospital, 5000 Odense, Denmark; (M.S.N.); (A.A.E.P.)
- Department of Clinical Research, Odense University Hospital, 5000 Odense, Denmark
| | - Anna C. Nilsson
- Department of Clinical Immunology, Odense University Hospital, 5000 Odense, Denmark;
| | - Justyna Okarmus
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (M.R.); (M.G.); (J.O.); (M.M.)
| | - Anne A. E. Poulsen
- Department of Neurology, Odense University Hospital, 5000 Odense, Denmark; (M.S.N.); (A.A.E.P.)
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (M.R.); (M.G.); (J.O.); (M.M.)
- Department of Neurology, Odense University Hospital, 5000 Odense, Denmark; (M.S.N.); (A.A.E.P.)
- Department of Clinical Research, Odense University Hospital, 5000 Odense, Denmark
- BRIDGE—Brain Research Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Morten Blaabjerg
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (M.R.); (M.G.); (J.O.); (M.M.)
- Department of Neurology, Odense University Hospital, 5000 Odense, Denmark; (M.S.N.); (A.A.E.P.)
- Department of Clinical Research, Odense University Hospital, 5000 Odense, Denmark
- BRIDGE—Brain Research Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
- Correspondence: ; Tel.: +45-2068-3863
| |
Collapse
|
13
|
Chakravarty D, Das Sarma J. Murine-β-coronavirus-induced neuropathogenesis sheds light on CNS pathobiology of SARS-CoV2. J Neurovirol 2021; 27:197-216. [PMID: 33547593 PMCID: PMC7864135 DOI: 10.1007/s13365-021-00945-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/29/2020] [Accepted: 01/12/2021] [Indexed: 02/06/2023]
Abstract
The pandemic caused by SARS-CoV-2 has caused widespread infection and significant mortality across the globe. Combined virology perspective of SARS-CoV-2 with a deep-rooted understanding of pathophysiological and immunological processes underlying the clinical manifestations of COVID-19 is of prime importance. The characteristic symptom of COVID-19 is respiratory distress with diffused alveolar damage, but emerging evidence suggests COVID-19 might also have neurologic consequences. Dysregulated homeostasis in the lungs has proven to be fatal, but one cannot ignore that the inability to breathe might be due to defects in the respiratory control center of the brainstem. While the mechanism of pulmonary distress has been documented in the literature, awareness of neurological features and their pathophysiology is still in the nascent state. This review makes references to the neuro-immune axis and neuro-invasive potential of SARS-CoV and SARS-CoV2, as well as the prototypic H-CoV strains in human brains. Simultaneously, considerable discussion on relevant experimental evidence of mild to severe neurological manifestations of fellow neurotropic murine-β-CoVs (m-CoVs) in the mouse model will help understand the underpinning mechanisms of Neuro-COVID. In this review, we have highlighted the neuroimmunopathological processes in murine CoVs. While MHV infection in mice and SARS-CoV-2 infection in humans share numerous parallels, there are critical differences in viral recognition and viral entry. These similarities are highlighted in this review, while differences have also been emphasized. Though CoV-2 Spike does not favorably interact with murine ACE2 receptor, modification of murine SARS-CoV2 binding domain or development of transgenic ACE-2 knock-in mice might help in mediating consequential infection and understanding human CoV2 pathogenesis in murine models. While a global animal model that can replicate all aspects of the human disease remains elusive, prior insights and further experiments with fellow m-β-CoV-induced cause-effect experimental models and current human COVID-19 patients data may help to mitigate the SARS-CoV-2-induced multifactorial multi-organ failure.
Collapse
Affiliation(s)
- Debanjana Chakravarty
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Haringhata, 741246, Mohanpur, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Haringhata, 741246, Mohanpur, India.
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
14
|
Das Sarma J, Burrows A, Rayman P, Hwang MH, Kundu S, Sharma N, Bergmann C, Sen GC. Ifit2 deficiency restricts microglial activation and leukocyte migration following murine coronavirus (m-CoV) CNS infection. PLoS Pathog 2020; 16:e1009034. [PMID: 33253295 PMCID: PMC7738193 DOI: 10.1371/journal.ppat.1009034] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/15/2020] [Accepted: 10/02/2020] [Indexed: 02/07/2023] Open
Abstract
The interferon-induced tetratricopeptide repeat protein (Ifit2) protects mice from lethal neurotropic viruses. Neurotropic coronavirus MHV-RSA59 infection of Ifit2-/- mice caused pronounced morbidity and mortality accompanied by rampant virus replication and spread throughout the brain. In spite of the higher virus load, induction of many cytokines and chemokines in the brains of infected Ifit2-/- mice were similar to that in wild-type mice. In contrast, infected Ifit2-/- mice revealed significantly impaired microglial activation as well as reduced recruitment of NK1.1 T cells and CD4 T cells to the brain, possibly contributing to the lack of viral clearance. These two deficiencies were associated with a lower level of microglial expression of CX3CR1, the receptor of the CX3CL1 (Fractalkine) chemokine, which plays a critical role in both microglial activation and leukocyte recruitment. The above results uncovered a new potential role of an interferon-induced protein in immune protection. Interferons (IFNs) are known to protect from virus dissemination and pathogenesis. Several IFN stimulated genes (ISG) regulate neuropathogenesis but the mechanisms underlying the antiviral effects are not clearly understood. IFN induced tetratricopeptide repeats (Ifit) are a class of ISGs. Among the Ifits, Ifit2 is known to play a beneficial role in restricting neurotropic viral replication. To provide better cellular insights into the protective mechanisms of Ifit2 functions, using a neurotropic coronavirus infection in Ifit2 depleted mice we report that in the absence of Ifit2, viral replication is dramatically increased and mice develop severe clinical signs and symptoms of neurological deficit. Despite the enormous viral load, Ifit2 deficient mice are impaired in microglial activation and recruitment of peripheral leukocytes into the CNS. This impaired leuocyte infiltration in Ifit2 deficient mice was also associated with reduced expression of a novel chemokine receptor CX3CR1,which is important for viral induced microglial activation and maintaining tissue homeostasis.
Collapse
Affiliation(s)
- Jayasri Das Sarma
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio, United States of America
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
- * E-mail:
| | - Amy Burrows
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio, United States of America
| | - Patricia Rayman
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio, United States of America
| | - Mi-Hyun Hwang
- Department of Neurosciences, Cleveland Clinic, Ohio, United States of America
| | - Soumya Kundu
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Nikhil Sharma
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio, United States of America
| | - Cornelia Bergmann
- Department of Neurosciences, Cleveland Clinic, Ohio, United States of America
| | - Ganes C. Sen
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio, United States of America
| |
Collapse
|