Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters

Database
Language
Journal subject
Affiliation country
Publication year range
1.
PLoS Pathog ; 15(12): e1008192, 2019 12.
Article in English | MEDLINE | ID: mdl-31809522

ABSTRACT

The hypoxia-inducible factor 1 alpha (HIF1α) protein and the hypoxic microenvironment are critical for infection and pathogenesis by the oncogenic gammaherpesviruses (γHV), Kaposi sarcoma herpes virus (KSHV) and Epstein-Barr virus (EBV). However, understanding the role of HIF1α during the virus life cycle and its biological relevance in the context of host has been challenging due to the lack of animal models for human γHV. To study the role of HIF1α, we employed the murine gammaherpesvirus 68 (MHV68), a rodent pathogen that readily infects laboratory mice. We show that MHV68 infection induces HIF1α protein and HIF1α-responsive gene expression in permissive cells. siRNA silencing or drug-inhibition of HIF1α reduce virus production due to a global downregulation of viral gene expression. Most notable was the marked decrease in many viral genes bearing hypoxia-responsive elements (HREs) such as the viral G-Protein Coupled Receptor (vGPCR), which is known to activate HIF1α transcriptional activity during KSHV infection. We found that the promoter of MHV68 ORF74 is responsive to HIF1α and MHV-68 RTA. Moreover, Intranasal infection of HIF1αLoxP/LoxP mice with MHV68 expressing Cre- recombinase impaired virus expansion during early acute infection and affected lytic reactivation in the splenocytes explanted from mice. Low oxygen concentrations accelerated lytic reactivation and enhanced virus production in MHV68 infected splenocytes. Thus, we conclude that HIF1α plays a critical role in promoting virus replication and reactivation from latency by impacting viral gene expression. Our results highlight the importance of the mutual interactions of the oxygen-sensing machinery and gammaherpesviruses in viral replication and pathogenesis.


Subject(s)
Gene Expression Regulation, Viral/physiology , Herpesviridae Infections/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Virus Latency/physiology , Virus Replication/physiology , Animals , Mice , Rhadinovirus/metabolism
2.
J Neuroinflammation ; 13(1): 125, 2016 05 31.
Article in English | MEDLINE | ID: mdl-27245318

ABSTRACT

BACKGROUND: Individuals suffering from spinal cord injury (SCI) are at higher risk for respiratory-related viral infections such as influenza. In a previous study (Zha et al., J Neuroinflammation 11:65, 2014), we demonstrated that chronic spinal cord injury caused impairment in CD8(+)T cell function with increased expression of the immunosuppressive protein, programmed cell death 1 (PD-1). The present study was undertaken to establish whether chronic SCI-induced immune deficits would affect antiviral immunity directed against primary and secondary infections. METHODS: Six to seven weeks following a SCI contusion at thoracic level T9, mice were infected intranasally with influenza virus. Virus-specific immunity was analyzed at various time points post-infection and compared to uninjured controls. RESULTS: We report that chronic thoracic SCI impairs the ability of the animals to mount an adequate antiviral immune response. While all uninjured control mice cleared the virus from their lungs by day 10 post-infection, a significant number (approximately 70 %) of chronic SCI mice did not clear the virus and succumbed to infection-induced mortality. This was attributed to severe deficits in both virus-specific antibody production and CD8(+) T cell response in injured mice after primary infection. We also determined that previously acquired humoral immunity was maintained after spinal cord injury as vaccination against influenza A prior to injury-protected mice from a homologous viral challenge. In contrast, prior immunization did not protect mice from a heterotypic challenge with a different strain of influenza virus. CONCLUSIONS: Taken together, our data demonstrate that chronic SCI attenuates virus-specific humoral and cellular immunity during the establishment of primary response and impairs the development of memory CD8(+) T cells. In contrast, B cell memory acquired through vaccination prior to SCI is preserved after injury which demonstrates that antigen-specific memory cells are refractory following injury. Our study defines important parameters of the deficits of chronic SCI-induced immune depression during a viral respiratory infection. Our objective is to better understand the mechanisms of spinal cord injury-induced immune depression with the goal of developing more effective therapies and reduce mortality due to complications from influenza and other infections.


Subject(s)
Antibodies, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular/physiology , Influenza A Virus, H3N2 Subtype/immunology , Spinal Cord Injuries/immunology , Animals , Antibodies, Viral/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Chronic Disease , Dogs , Female , Influenza A Virus, H3N2 Subtype/metabolism , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL , Orthomyxoviridae/immunology , Orthomyxoviridae/metabolism , Spinal Cord Injuries/metabolism , Thoracic Vertebrae
3.
Front Immunol ; 12: 705307, 2021.
Article in English | MEDLINE | ID: mdl-34512628

ABSTRACT

While apoptosis plays a role in B-cell self-tolerance, its significance in preventing autoimmunity remains unclear. Here, we report that dysregulated B cell apoptosis leads to delayed onset autoimmune phenotype in mice. Our longitudinal studies revealed that mice with B cell-specific deletion of pro-apoptotic Bim (BBimfl/fl ) have an expanded B cell compartment with a notable increase in transitional, antibody secreting and recently described double negative (DN) B cells. They develop greater hypergammaglobulinemia than mice lacking Bim in all cells and accumulate several autoantibodies characteristic of Systemic Lupus Erythematosus (SLE) and related Sjögren's Syndrome (SS) including anti-nuclear, anti-Ro/SSA and anti-La/SSB at a level comparable to NODH2h4 autoimmune mouse model. Furthermore, lymphocytes infiltrated the tissues including submandibular glands and formed follicle-like structures populated with B cells, plasma cells and T follicular helper cells indicative of ongoing immune reaction. This autoimmunity was ameliorated upon deletion of Bruton's tyrosine kinase (Btk) gene, which encodes a key B cell signaling protein. These studies suggest that Bim-mediated apoptosis suppresses and B cell tyrosine kinase signaling promotes B cell-mediated autoimmunity.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Apoptosis/physiology , Autoimmune Diseases/immunology , Autoimmunity/immunology , B-Lymphocytes/immunology , Bcl-2-Like Protein 11/physiology , Agammaglobulinaemia Tyrosine Kinase/deficiency , Agammaglobulinaemia Tyrosine Kinase/physiology , Animals , Antibody Specificity , Autoantibodies/blood , B-Lymphocytes/enzymology , B-Lymphocytes/pathology , Bcl-2-Like Protein 11/deficiency , Cell Division , Cells, Cultured , Hypergammaglobulinemia/immunology , Immune Tolerance/immunology , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen/immunology , Sjogren's Syndrome/immunology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL