Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 558
Filtrar
1.
J Neuroimmunol ; 393: 578382, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38850674

RESUMEN

Virus infections and autoimmune responses are implicated as primary triggers of demyelinating diseases. Specifically, the association of Epstein-Barr virus (EBV) infection with development of multiple sclerosis (MS) has re-ignited an interest in virus induced autoimmune responses to CNS antigens. Nevertheless, demyelination may also be caused by immune mediated bystander pathology in an attempt to control direct infection in the CNS. Tissue damage as a result of anti-viral responses or low level viral persistence may lead to immune activation manifesting in demyelinating lesions, axonal damage and clinical symptoms. This review focuses on the neurotropic mouse coronavirus induced demyelination model to highlight how immune responses activated during the acute phase pave the way to dampen pathology and promote repair. We specifically discuss the role of immune dampening factors programmed cell death ligand 1 (PD-L1) and interleukin (IL)-10, as well as microglia and triggering receptor expressed on myeloid cells 2 (Trem2), in limiting demyelination independent of viral persistence.


Asunto(s)
Enfermedades Desmielinizantes , Virus de la Hepatitis Murina , Animales , Ratones , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/virología , Enfermedades Desmielinizantes/patología , Enfermedades Desmielinizantes/etiología , Virus de la Hepatitis Murina/patogenicidad , Virus de la Hepatitis Murina/inmunología , Humanos , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/complicaciones , Modelos Animales de Enfermedad
2.
IUBMB Life ; 76(6): 313-331, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38116887

RESUMEN

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.


Asunto(s)
Antígenos CD40 , Ligando de CD40 , Enfermedades Desmielinizantes , Virus de la Hepatitis Murina , Enfermedades Neuroinflamatorias , Animales , Ligando de CD40/metabolismo , Ligando de CD40/genética , Ligando de CD40/inmunología , Enfermedades Neuroinflamatorias/patología , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/virología , Enfermedades Desmielinizantes/virología , Enfermedades Desmielinizantes/patología , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/metabolismo , Humanos , Antígenos CD40/metabolismo , Antígenos CD40/genética , Antígenos CD40/inmunología , Virus de la Hepatitis Murina/patogenicidad , Virus de la Hepatitis Murina/inmunología , Ratones , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/virología , Esclerosis Múltiple/patología , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Modelos Animales de Enfermedad
3.
Virology ; 566: 122-135, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34906793

RESUMEN

Mouse hepatitis virus (MHV; m-ß-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-ß-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-ß-CoV infected mice brain. Our study revealed that m-ß-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-ß-CoV-induced neuroinflammation.


Asunto(s)
Encéfalo/metabolismo , Infecciones por Coronavirus/metabolismo , Leucocitos/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Virus de la Hepatitis Murina/metabolismo , Enfermedades Neuroinflamatorias/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Animales , Encéfalo/virología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Virus de la Hepatitis Murina/inmunología , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/virología , Oxidación-Reducción , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba
4.
Science ; 373(6552)2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34103349

RESUMEN

The COVID-19 pandemic has revealed the pronounced vulnerability of the elderly and chronically ill to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced morbidity and mortality. Cellular senescence contributes to inflammation, multiple chronic diseases, and age-related dysfunction, but effects on responses to viral infection are unclear. Here, we demonstrate that senescent cells (SnCs) become hyper-inflammatory in response to pathogen-associated molecular patterns (PAMPs), including SARS-CoV-2 spike protein-1, increasing expression of viral entry proteins and reducing antiviral gene expression in non-SnCs through a paracrine mechanism. Old mice acutely infected with pathogens that included a SARS-CoV-2-related mouse ß-coronavirus experienced increased senescence and inflammation, with nearly 100% mortality. Targeting SnCs by using senolytic drugs before or after pathogen exposure significantly reduced mortality, cellular senescence, and inflammatory markers and increased antiviral antibodies. Thus, reducing the SnC burden in diseased or aged individuals should enhance resilience and reduce mortality after viral infection, including that of SARS-CoV-2.


Asunto(s)
Envejecimiento , Senescencia Celular/efectos de los fármacos , Infecciones por Coronavirus/mortalidad , Flavonoles/uso terapéutico , Moléculas de Patrón Molecular Asociado a Patógenos/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Animales , COVID-19/inmunología , COVID-19/mortalidad , Línea Celular , Infecciones por Coronavirus/inmunología , Dasatinib/farmacología , Dasatinib/uso terapéutico , Femenino , Flavonoles/farmacología , Regulación de la Expresión Génica , Humanos , Lipopolisacáridos , Masculino , Ratones , Ratones Endogámicos C57BL , Virus de la Hepatitis Murina/inmunología , Quercetina/farmacología , Quercetina/uso terapéutico , Receptores de Coronavirus/genética , Receptores de Coronavirus/metabolismo , Organismos Libres de Patógenos Específicos , Tratamiento Farmacológico de COVID-19
5.
Mediators Inflamm ; 2021: 9979032, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33967626

RESUMEN

Coronaviruses (CoVs) are enveloped and harbor an unusually large (30-32 kb) positive-strand linear RNA genome. Highly pathogenic coronaviruses cause severe acute respiratory syndrome (SARS) (SARS-CoV and SARS-CoV-2) and Middle East respiratory syndrome (MERS) (MERS-CoV) in humans. The coronavirus mouse hepatitis virus (MHV) infects mice and serves as an ideal model of viral pathogenesis, mainly because experiments can be conducted using animal-biosafety level-2 (A-BSL2) containment. Human thymosin beta-4 (Tß4), a 43-residue peptide with an acetylated N-terminus, is widely expressed in human tissues. Tß4 regulates actin polymerization and functions as an anti-inflammatory molecule and an antioxidant as well as a promoter of wound healing and angiogenesis. These activities led us to test whether Tß4 serves to treat coronavirus infections of humans. To test this possibility, here, we established a BALB/c mouse model of coronavirus infection using mouse CoV MHV-A59 to evaluate the potential protective effect of recombinant human Tß4 (rhTß4). Such a system can be employed under A-BSL2 containment instead of A-BSL3 that is required to study coronaviruses infectious for humans. We found that rhTß4 significantly increased the survival rate of mice infected with MHV-A59 through inhibiting virus replication, balancing the host's immune response, alleviating pathological damage, and promoting repair of the liver. These results will serve as the basis for further application of rhTß4 to the treatment of human CoV diseases such as COVID-19.


Asunto(s)
Infecciones por Coronavirus/tratamiento farmacológico , Virus de la Hepatitis Murina , Timosina/uso terapéutico , Animales , Anticuerpos Antivirales/sangre , Proteína C-Reactiva/análisis , Citocinas/sangre , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Virus de la Hepatitis Murina/inmunología , ARN Viral/análisis , Proteínas Recombinantes/uso terapéutico , Replicación Viral/efectos de los fármacos
6.
Nat Immunol ; 22(4): 510-519, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33707780

RESUMEN

Fibroblastic reticular cells (FRCs) determine the organization of lymphoid organs and control immune cell interactions. While the cellular and molecular mechanisms underlying FRC differentiation in lymph nodes and the splenic white pulp have been elaborated to some extent, in Peyer's patches (PPs) they remain elusive. Using a combination of single-cell transcriptomics and cell fate mapping in advanced mouse models, we found that PP formation in the mouse embryo is initiated by an expansion of perivascular FRC precursors, followed by FRC differentiation from subepithelial progenitors. Single-cell transcriptomics and cell fate mapping confirmed the convergence of perivascular and subepithelial FRC lineages. Furthermore, lineage-specific loss- and gain-of-function approaches revealed that the two FRC lineages synergistically direct PP organization, maintain intestinal microbiome homeostasis and control anticoronavirus immune responses in the gut. Collectively, this study reveals a distinct mosaic patterning program that generates key stromal cell infrastructures for the control of intestinal immunity.


Asunto(s)
Linaje de la Célula , Fibroblastos/inmunología , Inmunidad Mucosa , Mucosa Intestinal/inmunología , Intestino Delgado/inmunología , Ganglios Linfáticos Agregados/inmunología , Animales , Comunicación Celular , Células Cultivadas , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/virología , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Microbioma Gastrointestinal , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Interacciones Huésped-Patógeno , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/virología , Intestino Delgado/metabolismo , Intestino Delgado/microbiología , Intestino Delgado/virología , Ratones Endogámicos C57BL , Ratones Noqueados , Virus de la Hepatitis Murina/inmunología , Virus de la Hepatitis Murina/patogenicidad , Ganglios Linfáticos Agregados/metabolismo , Ganglios Linfáticos Agregados/microbiología , Ganglios Linfáticos Agregados/virología , Fenotipo , Análisis de la Célula Individual , Transcriptoma
7.
Eur J Immunol ; 51(5): 1062-1070, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33687066

RESUMEN

Coronaviruses (CoVs) represent enveloped, ss RNA viruses with the ability to infect a range of vertebrates causing mainly lung, CNS, enteric, and hepatic disease. While the infection with human CoV is commonly associated with mild respiratory symptoms, the emergence of SARS-CoV, MERS-CoV, and SARS-CoV-2 highlights the potential for CoVs to cause severe respiratory and systemic disease. The devastating global health burden caused by SARS-CoV-2 has spawned countless studies seeking clinical correlates of disease severity and host susceptibility factors, revealing a complex network of antiviral immune circuits. The mouse hepatitis virus (MHV) is, like SARS-CoV-2, a beta-CoV and is endemic in wild mice. Laboratory MHV strains have been extensively studied to reveal coronavirus virulence factors and elucidate host mechanisms of antiviral immunity. These are reviewed here with the aim to identify translational insights for SARS-CoV-2 learned from murine CoVs.


Asunto(s)
Inmunidad Adaptativa/inmunología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/patología , Virus de la Hepatitis Murina/inmunología , Virus de la Hepatitis Murina/patogenicidad , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , SARS-CoV-2/inmunología , Índice de Severidad de la Enfermedad , Glicoproteína de la Espiga del Coronavirus/metabolismo , Tropismo Viral/fisiología
8.
J Neurovirol ; 27(2): 197-216, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33547593

RESUMEN

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-ß-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-ß-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.


Asunto(s)
COVID-19/patología , Modelos Animales de Enfermedad , Virus de la Hepatitis Murina/patogenicidad , Neuroinmunomodulación/fisiología , Animales , COVID-19/inmunología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/patología , Humanos , Ratones , Virus de la Hepatitis Murina/inmunología , SARS-CoV-2
9.
J Am Assoc Lab Anim Sci ; 60(2): 152-159, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33500021

RESUMEN

Sentinel animals remain a common means of evaluating rodent health in research colonies. An evaluation of our sentinel program revealed that using Crl:CD1(ICR)-Elite (CD1-E) mice was expensive, occasionally disrupted by limited supply, and minimally responsive to the 3Rs. This evaluation prompted us to explore the use of CRL:NU-Foxn1nu/+ (Het-nude) mice as soiled-bedding sentinel (SBS) animals. Het-nude mice are a byproduct of breeding outbred athymic nude mice and are reared in isolators, with similar health status as CD1-E. Het-nude mice have a thymus, but may have smaller thymic size and fewer bone marrow stem cells than do wildtype controls, suggesting that Het-nude mice might not be immunologically normal. This study compared the antibody titer and seroconversion kinetics of Het-nude and CD1-E SBS to murine norovirus (MNV) and mouse hepatitis virus (MHV). Het-nude and CD1-E female SBS (n = 22 mice of each stock) were housed continuously on soiled bedding collected from MNV-positive or MNV- and MHV-positive colonies at cage changes. Blood was collected for serology at 3, 9 and 12 to 19 wk after the start of soiled bedding exposure. Antibody titers to MNV or MHV did not differ significantly between Het-nude and CD1-E mice. A significant relationship was found between weeks of exposure and titer levels with an increase in titer over the testing period. This study supports the possible use of Het-nude mice as SBS, given that their antibody responses to MNV and MHV are equivalent to those of CD1-E mice.


Asunto(s)
Virus de la Hepatitis Murina/inmunología , Norovirus/inmunología , Enfermedades de los Roedores/virología , Animales , Ropa de Cama y Ropa Blanca , Femenino , Vivienda para Animales , Cinética , Ratones , Ratones Desnudos , Enfermedades de los Roedores/sangre , Enfermedades de los Roedores/inmunología , Vigilancia de Guardia , Seroconversión
10.
J Immunol ; 205(12): 3372-3382, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33188072

RESUMEN

Persistent infection with gammaherpesviruses (γHV) can cause lymphomagenesis in immunocompromised patients. Murine γHV-68 (MHV-68) is an important tool for understanding immune factors contributing to γHV control; however, modeling control of γHV-associated lymphomagenesis has been challenging. Current model systems require very long incubation times or severe immune suppression, and tumor penetrance is low. In this report, we describe the generation of a B cell lymphoma on the C57BL/6 background, which is driven by the Myc oncogene and expresses an immunodominant CD8 T cell epitope from MHV-68. We determined MHV-68-specific CD8 T cells in latently infected mice use either IFN-γ or perforin/granzyme to control γHV-associated lymphoma, but perforin/granzyme is a more potent effector mechanism for lymphoma control than IFN-γ. Consistent with previous reports, CD4-depleted mice lost control of virus replication in persistently infected mice. However, control of lymphoma remained intact in the absence of CD4 T cells. Collectively, these data show the mechanisms of T cell control of B cell lymphoma in γHV-infected mice overlap with those necessary for control of virus replication, but there are also important differences. This study establishes a tool for further dissecting immune surveillance against, and optimizing adoptive T cell therapies for, γHV-associated lymphomas.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Memoria Inmunológica , Linfoma de Células B/inmunología , Virus de la Hepatitis Murina/inmunología , Proteínas de Neoplasias/inmunología , Animales , Epítopos de Linfocito T/genética , Femenino , Linfoma de Células B/genética , Linfoma de Células B/patología , Ratones , Ratones Transgénicos , Virus de la Hepatitis Murina/genética , Proteínas de Neoplasias/genética
11.
Glia ; 68(11): 2345-2360, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32449994

RESUMEN

The present study examines functional contributions of microglia in host defense, demyelination, and remyelination following infection of susceptible mice with a neurotropic coronavirus. Treatment with PLX5622, an inhibitor of colony stimulating factor 1 receptor (CSF1R) that efficiently depletes microglia, prior to infection of the central nervous system (CNS) with the neurotropic JHM strain of mouse hepatitis virus (JHMV) resulted in increased mortality compared with control mice that correlated with impaired control of viral replication. Single cell RNA sequencing (scRNASeq) of CD45+ cells isolated from the CNS revealed that PLX5622 treatment resulted in muted CD4+ T cell activation profile that was associated with decreased expression of transcripts encoding MHC class II and CD86 in macrophages but not dendritic cells. Evaluation of spinal cord demyelination revealed a marked increase in white matter damage in PLX5622-treated mice that corresponded with elevated expression of transcripts encoding disease-associated proteins Osteopontin (Spp1), Apolipoprotein E (Apoe), and Triggering receptor expressed on myeloid cells 2 (Trem2) that were enriched within macrophages. In addition, PLX5622 treatment dampened expression of Cystatin F (Cst7), Insulin growth factor 1 (Igf1), and lipoprotein lipase (Lpl) within macrophage populations which have been implicated in promoting repair of damaged nerve tissue and this was associated with impaired remyelination. Collectively, these findings argue that microglia tailor the CNS microenvironment to enhance control of coronavirus replication as well as dampen the severity of demyelination and influence repair.


Asunto(s)
Encéfalo/inmunología , Infecciones por Coronavirus/inmunología , Interacciones Huésped-Patógeno/inmunología , Microglía/inmunología , Virus de la Hepatitis Murina/inmunología , Compuestos Orgánicos/toxicidad , Animales , Encéfalo/efectos de los fármacos , Encéfalo/virología , Infecciones por Coronavirus/inducido químicamente , Interacciones Huésped-Patógeno/efectos de los fármacos , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/virología
12.
Exp Mol Pathol ; 115: 104474, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32454103

RESUMEN

The pathogenesis of viral infections involves an immune response by cytokines, causing a deleterious effect on organ function, in addition to tissue destruction due to viral replication. Clinical symptoms and laboratory findings of the human coronavirus disease COVID-19, caused by the novel coronavirus SARS CoV-2, indicate cytokine involvement. Our laboratory showed that an experimental murine coronavirus (MHV-A59) can be transmitted into the brain by intranasal or intracerebral exposure and that neurovirulence is mediated by cytokine secretion. In this study we investigated which cells in the brain produce cytokines, thus functioning as the brain's innate immune system. Using tissue cultures of microglia, and clonal populations of astrocytes, we found that microglia and type I astrocytes (but not types II and III), produced pro-inflammatory cytokines in response to MHV-A59 infection. A molecularly closely related, non-encephalitic strain of the virus (MHV-2) caused in vitro infection, but without cytokine induction. Furthermore, immunofluorescence and immunohistochemistry revealed that type I astrocytes and microglia have perivascular foot processes necessary for the formation of the perivascular glymphatic system, the anatomical site of the brain's innate immune system. Cytokine secretion by type I astrocytes and microglia, as part of the brain's glymphatic and innate immune system, contributes to the pathogenesis of an encephalitic coronavirus infection, and indicates the rationale for anti-cytokine therapies for COVID-19.


Asunto(s)
Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/transmisión , Virus de la Hepatitis Murina/metabolismo , Animales , Astrocitos/inmunología , Betacoronavirus , Encéfalo/inmunología , Encéfalo/patología , COVID-19 , Línea Celular , Células Cultivadas , Coronavirus/metabolismo , Infecciones por Coronavirus/virología , Citocinas/inmunología , Humanos , Ratones , Microglía/inmunología , Virus de la Hepatitis Murina/inmunología , Virus de la Hepatitis Murina/patogenicidad , Pandemias , Neumonía Viral , SARS-CoV-2 , Replicación Viral/inmunología , Replicación Viral/fisiología
13.
Immunol Lett ; 217: 25-30, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31726186

RESUMEN

In a previous work we demonstrated that inhibition of mouse indoleamine 2,3-dioxygenase (IDO) by methyltryptophan (MT) exacerbated the pathological actions of mouse hepatitis virus (MHV-A59) infection, suggesting that tryptophan (TRP) catabolism was involved in viral effects. Since there is a second enzyme that dioxygenates TRP, tryptophan-2, 3-dioxygenase (TDO), which is mainly located in liver, we decided to study its role in our model of MHV-infection. Results showed that in vivo TDO inhibition by LM10, a derivative of 3-(2-(pyridyl) ethenyl) indole, resulted in a decrease of anti- MHV Ab titers induced by the virus infection. Besides, a reduction of some alarmin release, i.e, uric acid and high-mobility group box1 protein (HMGB1), was observed. Accordingly, since alarmin liberation was related to the expression of autoantibodies (autoAb) to fumarylacetoacetate hydrolase (FAH), these autoAb also diminished. Moreover, PCR results indicated that TDO inhibition did not abolish viral replication. Furthermore, histological liver examination did not reveal strong pathologies, whereas mouse survival was hundred percent in control as well as in MHV-infected mice treated with LM10. Data presented in this work indicate that in spite of the various TDO actions already described, specific TDO blockage could also restrain some MHV actions, mainly suppressing autoimmune reactions. Such results should prompt further experiments with various viruses to confirm the possible use of a TDO inhibitor such as LM-10 to treat either viral infections or even autoimmune diseases triggered by a viral infection.


Asunto(s)
Enfermedades Autoinmunes/enzimología , Autoinmunidad/efectos de los fármacos , Infecciones por Coronavirus/enzimología , Infecciones por Coronavirus/inmunología , Hígado/enzimología , Virus de la Hepatitis Murina/inmunología , Triptófano Oxigenasa/antagonistas & inhibidores , Triptófano Oxigenasa/metabolismo , Alarminas/metabolismo , Animales , Autoanticuerpos/efectos de los fármacos , Autoanticuerpos/inmunología , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/virología , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Femenino , Proteína HMGB1/sangre , Proteína HMGB1/metabolismo , Hidrolasas/inmunología , Indoles/uso terapéutico , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/patología , Ratones , Ratones Endogámicos BALB C , Virus de la Hepatitis Murina/efectos de los fármacos , Virus de la Hepatitis Murina/crecimiento & desarrollo , Triptófano/metabolismo , Triptófano Oxigenasa/genética , Ácido Úrico/sangre , Ácido Úrico/metabolismo , Replicación Viral/efectos de los fármacos , Replicación Viral/inmunología
14.
Front Immunol ; 10: 2473, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31736943

RESUMEN

Calcium (Ca2+) is a vital secondary messenger in T lymphocytes regulating a vast array of important events including maturation, homeostasis, activation, and apoptosis and can enter the cell through CRAC, TRP, and CaV channels. Here we describe a mutation in the L-type Ca2+ channel CaV1.4 leading to T lymphocyte dysfunction, including several hallmarks of immunological exhaustion. CaV1.4-deficient mice exhibited an expansion of central and effector memory T lymphocytes, and an upregulation of inhibitory receptors on several T cell subsets. Moreover, the sustained elevated levels of activation markers on B lymphocytes suggest that they are in a chronic state of activation. Functionally, T lymphocytes exhibited a reduced store-operated Ca2+ flux compared to wild-type controls. Finally, modifying environmental conditions by herpes virus infection exacerbated the dysfunctional immune phenotype of the CaV1.4-deficient mice. This is the first example where the mutation of a CaV channel leads to T lymphocyte dysfunction, including the upregulation of several inhibitory receptors, hallmarks of T cell exhaustion, and establishes the physiological importance of CaV channel signaling in maintaining a nimble immune system.


Asunto(s)
Canales de Calcio Tipo L/genética , Mutación , Fenotipo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Relación CD4-CD8 , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Calcio/metabolismo , Señalización del Calcio , Expresión Génica , Estudios de Asociación Genética , Hepatitis Viral Animal/inmunología , Hepatitis Viral Animal/virología , Memoria Inmunológica , Inmunofenotipificación , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Recuento de Linfocitos , Ratones , Ratones Noqueados , Virus de la Hepatitis Murina/inmunología
15.
Viral Immunol ; 32(1): 25-37, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30109979

RESUMEN

Chemokines (chemotactic cytokines) are involved in a wide variety of biological processes. Following microbial infection, there is often robust chemokine signaling elicited from infected cells, which contributes to both innate and adaptive immune responses that control growth of the invading pathogen. Infection of the central nervous system (CNS) by the neuroadapted John Howard Mueller (JHM) strain of mouse hepatitis virus (JHMV) provides an excellent example of how chemokines aid in host defense as well as contribute to disease. Intracranial inoculation of the CNS of susceptible mice with JHMV results in an acute encephalomyelitis characterized by widespread dissemination of virus throughout the parenchyma. Virus-specific T cells are recruited to the CNS, and control viral replication through release of antiviral cytokines and cytolytic activity. Sterile immunity is not acquired, and virus will persist primarily in white matter tracts leading to chronic neuroinflammation and demyelination. Chemokines are expressed and contribute to defense as well as chronic disease by attracting targeted populations of leukocytes to the CNS. The T cell chemoattractant chemokine CXCL10 (interferon-inducible protein 10 kDa, IP-10) is prominently expressed in both stages of disease, and serves to attract activated T and B lymphocytes expressing CXC chemokine receptor 3 (CXCR3), the receptor for CXCL10. Functional studies that have blocked expression of either CXCL10 or CXCR3 illuminate the important role of this signaling pathway in host defense and neurodegeneration in a model of viral-induced neurologic disease.


Asunto(s)
Enfermedades Virales del Sistema Nervioso Central/inmunología , Quimiocina CXCL10/inmunología , Infecciones por Coronavirus/inmunología , Enfermedades Desmielinizantes/inmunología , Virus de la Hepatitis Murina/inmunología , Animales , Enfermedades Virales del Sistema Nervioso Central/patología , Quimiocina CXCL10/genética , Factores Quimiotácticos , Infecciones por Coronavirus/patología , Enfermedades Desmielinizantes/virología , Humanos , Ratones , Ratones Endogámicos C57BL , Virus de la Hepatitis Murina/patogenicidad , Receptores CXCR3/genética , Receptores CXCR3/inmunología , Transducción de Señal , Linfocitos T/inmunología
16.
JCI Insight ; 3(11)2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29875310

RESUMEN

The nasal mucosa is an important component of mucosal immunity. Immunogenic particles in inspired air are known to activate the local nasal mucosal immune system and can lead to sinonasal inflammation; however, little is known about the effect of this activation on the lung immune environment. Here, we showed that nasal inoculation of murine coronavirus (CoV) in the absence of direct lung infection primes the lung immune environment by recruiting activated monocytes (Ly6C+ inflammatory monocytes) and NK cells into the lungs. Unlike infiltration of these cells into directly infected lungs, a process that requires type I IFN signaling, nasally induced infiltration of Ly6C+ inflammatory monocytes into the lungs is IFN-I independent. These activated macrophages ingested antigen and migrated to pulmonary lymph nodes, and enhanced both innate and adaptive immunity after heterologous virus infection. Clinically, such nasal-only inoculation of MHV-1 failed to cause pneumonia but significantly reduced mortality and morbidity of lethal pneumonia caused by severe acute respiratory syndrome CoV (SARS-CoV) or influenza A virus. Together, the data indicate that the nose and upper airway remotely prime the lung immunity to protect the lungs from direct viral infections.


Asunto(s)
Virus de la Hepatitis Murina/inmunología , Mucosa Nasal/inmunología , Neumonía Viral/inmunología , Vacunas contra Hepatitis Viral/administración & dosificación , Administración Intranasal , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunidad Innata , Inmunidad Mucosa , Virus de la Influenza A/inmunología , Virus de la Influenza A/patogenicidad , Pulmón/citología , Pulmón/inmunología , Pulmón/virología , Activación de Macrófagos , Macrófagos/inmunología , Ratones , Mucosa Nasal/citología , Mucosa Nasal/virología , Neumonía Viral/mortalidad , Neumonía Viral/prevención & control , Neumonía Viral/virología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad
17.
J Virol ; 92(10)2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29491163

RESUMEN

The contribution of distinct central nervous system (CNS) resident cells to protective alpha/beta interferon (IFN-α/ß) function following viral infections is poorly understood. Based on numerous immune regulatory functions of astrocytes, we evaluated the contribution of astrocyte IFN-α/ß signaling toward protection against the nonlethal glia- and neuronotropic mouse hepatitis virus (MHV) strain A59. Analysis of gene expression associated with IFN-α/ß function, e.g., pattern recognition receptors (PRRs) and interferon-stimulated genes (ISGs), revealed lower basal mRNA levels in brain-derived astrocytes than in microglia. Although astrocytes poorly induced Ifnß mRNA following infection, they upregulated various mRNAs in the IFN-α/ß pathway to a higher extent than microglia, supporting effective IFN-α/ß responsiveness. Ablation of the IFN-α/ß receptor (IFNAR) in astrocytes using mGFAPcre IFNARfl/fl mice resulted in severe encephalomyelitis and mortality, coincident with uncontrolled virus replication. Further, virus spread was not restricted to astrocytes but also affected microglia and neurons, despite increased and sustained Ifnα/ß and ISG mRNA levels within the CNS. IFN-γ, a crucial mediator for MHV control, was not impaired in infected mGFAPcre IFNARfl/fl mice despite reduced T cell CNS infiltration. Unexpectedly however, poor induction of IFN-γ-dependent major histocompatibility complex (MHC) class II expression on microglia supported that defective IFN-γ signaling contributes to uncontrolled virus replication. A link between sustained elevated IFN-α/ß and impaired responsiveness to IFN-γ supports the novel concept that temporally limited early IFN-α/ß responses are critical for effective antiviral IFN-γ function. Overall, our results imply that IFN-α/ß signaling in astrocytes is not only critical in limiting early CNS viral spread but also promotes protective antiviral IFN-γ function.IMPORTANCE An antiviral state established by IFN-α/ß contains initial viral spread as adaptive immunity develops. While it is apparent that the CNS lacks professional IFN-α/ß producers and that resident cells have distinct abilities to elicit innate IFN-α/ß responses, protective interactions between inducer and responder cells require further investigation. Infection with a glia- and neuronotropic coronavirus demonstrates that astrocytes mount a delayed but more robust response to infection than microglia, despite their lower basal mRNA levels of IFN-α/ß-inducing components. Lethal, uncontrolled viral dissemination following ablation of astrocyte IFN-α/ß signaling revealed the importance of IFN-α/ß responses in a single cell type for protection. Sustained global IFN-α/ß expression associated with uncontrolled virus did not suffice to protect neurons and further impaired responsiveness to protective IFN-γ. The results support astrocytes as critical contributors to innate immunity and the concept that limited IFN-α/ß responses are critical for effective subsequent antiviral IFN-γ function.


Asunto(s)
Astrocitos/inmunología , Interferón-alfa/inmunología , Interferón beta/inmunología , Interferón gamma/inmunología , Virus de la Hepatitis Murina/inmunología , Receptor de Interferón alfa y beta/genética , Animales , Astrocitos/virología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/virología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Encefalomielitis/inmunología , Encefalomielitis/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/inmunología
18.
Front Immunol ; 9: 2935, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30619295

RESUMEN

Background: Fulminant hepatitis (FH) is a serious threat to human life, accompanied by massive and rapid necroinflammation. Kupffer cells, the major immune cell population involved in innate immune responses, are considered to be central for FH. Fibrinogen-like protein 2 (Fgl2) is a pro-coagulant protein that is substantially induced in macrophages upon viral infection, and Fgl2 depletion represses murine hepatitis virus strain 3 (MHV-3) infection. Clara cell 10 kDa (CC10) protein is a secretory protein with anti-inflammatory properties in allergic rhinitis and asthma. However, its mechanisms of action and pathogenic roles in other disease are still unclear. In this study, we aimed to determine the role of CC10 in FH and the regulation of Fgl2 by CC10. Methods: A mouse FH model was established by peritoneal injection of MHV-3. The mice received CC10 protein through tail vein injection before viral infection. Survival rate, liver function, liver histology, fibrin deposition, and necrosis were examined. The regulatory effect of CC10 on Fgl2 expression was investigated using THP-1 cells and mouse peritoneal macrophages in vitro. Results: In the mouse FH model induced by MHV-3, the survival rate increased from 0 to 12.5% in the CC10 group compared to that in the saline-only control group. Meanwhile, the levels of ALT and AST in serum were significantly decreased and liver damage was reduced. Furthermore, hepatic Fgl2, TNF-α, and IL-1ß expression was obviously downregulated together with fibrin deposition, and hepatocyte apoptosis was reduced after administration of CC10 protein. In vitro, CC10 was found to significantly inhibit the expression of Fgl2 in IFN-γ-treated THP-1 cells and MHV-3-infected mouse peritoneal macrophages by western blot and real-time PCR. However, there was no direct interaction between CC10 and Fgl2 as shown by co-immunoprecipitation. Microarray investigations suggested that HMG-box transcription factor 1 (HBP1) was significantly low in CC10-treated and IFN-γ-primed THP-1 cells. HBP1-siRNA treatment abrogated the inhibitory effect of CC10 on Fgl2 expression in Human Umbilical Vein Endothelial cells (HUVECs). Conclusion:CC10 protects against MHV-3-induced FH via suppression of Fgl2 expression in macrophages. Such effects may be mediated by the transcription factor HBP1.


Asunto(s)
Infecciones por Coronavirus/inmunología , Fibrinógeno/metabolismo , Hepatitis Viral Animal/inmunología , Fallo Hepático Agudo/inmunología , Uteroglobina/metabolismo , Animales , Células CHO , Infecciones por Coronavirus/mortalidad , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Cricetulus , Modelos Animales de Enfermedad , Femenino , Fibrinógeno/genética , Hepatitis Viral Animal/mortalidad , Hepatitis Viral Animal/patología , Hepatitis Viral Animal/virología , Proteínas del Grupo de Alta Movilidad/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hígado/inmunología , Hígado/patología , Hígado/virología , Fallo Hepático Agudo/mortalidad , Fallo Hepático Agudo/patología , Fallo Hepático Agudo/virología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Virus de la Hepatitis Murina/inmunología , Virus de la Hepatitis Murina/patogenicidad , Necrosis/inmunología , Necrosis/patología , Necrosis/virología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Represoras/metabolismo , Tasa de Supervivencia , Células THP-1 , Uteroglobina/genética
19.
Front Immunol ; 9: 3022, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30619363

RESUMEN

The central nervous system (CNS) is vulnerable to several viral infections including herpes viruses, arboviruses and HIV to name a few. While a rapid and effective immune response is essential to limit viral spread and mortality, this anti-viral response needs to be tightly regulated in order to limit immune mediated tissue damage. This balance between effective virus control with limited pathology is especially important due to the highly specialized functions and limited regenerative capacity of neurons, which can be targets of direct virus cytolysis or bystander damage. CNS infection with the neurotropic strain of mouse hepatitis virus (MHV) induces an acute encephalomyelitis associated with focal areas of demyelination, which is sustained during viral persistence. Both innate and adaptive immune cells work in coordination to control virus replication. While type I interferons are essential to limit virus spread associated with early mortality, perforin, and interferon-γ promote further virus clearance in astrocytes/microglia and oligodendrocytes, respectively. Effective control of virus replication is nonetheless associated with tissue damage, characterized by demyelinating lesions. Interestingly, the anti-inflammatory cytokine IL-10 limits expansion of tissue lesions during chronic infection without affecting viral persistence. Thus, effective coordination of pro- and anti-inflammatory cytokines is essential during MHV induced encephalomyelitis in order to protect the host against viral infection at a limited cost.


Asunto(s)
Infecciones por Coronavirus/inmunología , Encefalomielitis/inmunología , Interferones/inmunología , Interleucina-10/inmunología , Virus de la Hepatitis Murina/inmunología , Animales , Sistema Nervioso Central/inmunología , Infecciones por Coronavirus/virología , Modelos Animales de Enfermedad , Encefalomielitis/virología , Interacciones Huésped-Patógeno/inmunología , Interferones/metabolismo , Interleucina-10/metabolismo , Ratones , Perforina/inmunología , Perforina/metabolismo
20.
J Virol ; 92(1)2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29046453

RESUMEN

Coronaviruses (CoVs) are positive-sense RNA viruses that infect numerous mammalian and avian species and are capable of causing severe and lethal disease in humans. CoVs encode several innate immune antagonists that counteract the host innate immune response to facilitate efficient viral replication. CoV nonstructural protein 14 (nsp14) encodes 3'-to-5' exoribonuclease activity (ExoN), which performs a proofreading function and is required for high-fidelity replication. Outside of the order Nidovirales, arenaviruses are the only RNA viruses that encode an ExoN, which functions to degrade double-stranded RNA (dsRNA) replication intermediates. In this study, we tested the hypothesis that CoV ExoN also functions to antagonize the innate immune response. We demonstrate that viruses lacking ExoN activity [ExoN(-)] are sensitive to cellular pretreatment with interferon beta (IFN-ß) in a dose-dependent manner. In addition, ExoN(-) virus replication was attenuated in wild-type bone marrow-derived macrophages (BMMs) and partially restored in interferon alpha/beta receptor-deficient (IFNAR-/-) BMMs. ExoN(-) virus replication did not result in IFN-ß gene expression, and in the presence of an IFN-ß-mediated antiviral state, ExoN(-) viral RNA levels were not substantially reduced relative to those of untreated samples. However, ExoN(-) virus generated from IFN-ß-pretreated cells had reduced specific infectivity and decreased relative fitness, suggesting that ExoN(-) virus generated during an antiviral state is less viable to establish a subsequent infection. Overall, our data suggest murine hepatitis virus (MHV) ExoN activity is required for resistance to the innate immune response, and antiviral mechanisms affecting the viral RNA sequence and/or an RNA modification act on viruses lacking ExoN activity.IMPORTANCE CoVs encode multiple antagonists that prevent or disrupt an efficient innate immune response. Additionally, no specific antiviral therapies or vaccines currently exist for human CoV infections. Therefore, the study of CoV innate immune antagonists is essential for understanding how CoVs overcome host defenses and to maximize potential therapeutic interventions. Here, we sought to determine the contributions of nsp14 ExoN activity in the induction of and resistance to the innate immune response. We show that viruses lacking nsp14 ExoN activity are more sensitive than wild-type MHV to restriction by exogenous IFN-ß and that viruses produced in the presence of an antiviral state are less capable of establishing a subsequent viral infection. Our results support the hypothesis that murine hepatitis virus ExoN activity is required for resistance to the innate immune response.


Asunto(s)
Exorribonucleasas/genética , Exorribonucleasas/metabolismo , Inmunidad Innata , Virus de la Hepatitis Murina/enzimología , Virus de la Hepatitis Murina/inmunología , Proteínas no Estructurales Virales/metabolismo , Animales , Antivirales/farmacología , Genoma Viral , Interferón beta/farmacología , Ratones , Virus de la Hepatitis Murina/efectos de los fármacos , Virus de la Hepatitis Murina/genética , Mutagénesis , Mutación , ARN Viral/metabolismo , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...