Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(36): 18001-18008, 2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31427525

RESUMO

Lymphocytic choriomeningitis virus (LCMV) WE variant 2.2 (v2.2) generated a high level of the major mouse urinary protein: MUP. Mice infected with LCMV WE v54, which differed from v2.2 by a single amino acid in the viral glycoprotein, failed to generate MUP above baseline levels found in uninfected controls. Variant 54 bound at 2.5 logs higher affinity to the LCMV receptor α-dystroglycan (α-DG) than v2.2 and entered α-DG-expressing but not α-DG-null cells. Variant 2.2 infected both α-DG-null or -expressing cells. Variant 54 infected more dendritic cells, generated a negligible CD8 T cell response, and caused a persistent infection, while v2.2 generated cytotoxic T lymphocytes (CTLs) and cleared virus within 10 days. By 20 days postinfection and through the 80-day observation period, significantly higher amounts of MUP were found in v2.2-infected mice. Production of MUP was dependent on virus-specific CTL as deletion of such cells aborted MUP production. Furthermore, MUP production was not elevated in v2.2 persistently infected mice unless virus was cleared following transfer of virus-specific CTL.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Regulação da Expressão Gênica/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Proteínas/imunologia , Animais , Distroglicanas/imunologia , Coriomeningite Linfocítica/patologia , Camundongos
2.
Int J Mol Sci ; 23(5)2022 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-35269842

RESUMO

Mycobacterium tuberculosis (Mtb) represents a major burden to global health, and refined vaccines are needed. Replication-deficient lymphocytic choriomeningitis virus (rLCMV)-based vaccine vectors against cytomegalovirus have proven safe for human use and elicited robust T cell responses in a large proportion of vaccine recipients. Here, we developed an rLCMV vaccine expressing the Mtb antigens TB10.4 and Ag85B. In mice, rLCMV elicited high frequencies of polyfunctional Mtb-specific CD8 and CD4 T cell responses. CD8 but not CD4 T cells were efficiently boosted upon vector re-vaccination. High-frequency responses were also observed in neonatally vaccinated mice, and co-administration of rLCMV with Expanded Program of Immunization (EPI) vaccines did not result in substantial reciprocal interference. Importantly, rLCMV immunization significantly reduced the lung Mtb burden upon aerosol challenge, resulting in improved lung ventilation. Protection was associated with increased CD8 T cell recruitment but reduced CD4 T cell infiltration upon Mtb challenge. When combining rLCMV with BCG vaccination in a heterologous prime-boost regimen, responses to the rLCMV-encoded Mtb antigens were further augmented, but protection was not significantly different from rLCMV or BCG vaccination alone. This work suggests that rLCMV may show utility for neonatal and/or adult vaccination efforts against pulmonary tuberculosis.


Assuntos
Mycobacterium tuberculosis , Animais , Antígenos de Bactérias , Vacina BCG , Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos , Vírus da Coriomeningite Linfocítica/genética , Camundongos , Mycobacterium tuberculosis/genética
3.
Eur J Immunol ; 48(8): 1389-1399, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29684247

RESUMO

Tr1 cell therapy is considered an emerging approach to improve transplant tolerance and enhance allogeneic graft survival. However, it remains unclear how Tr1 cells promote transplant tolerance and whether they will be safe and stable in the face of an acute viral infection. By employing a mouse model of pancreatic islet transplantation, we report that Tr1 cell therapy promoted transplant tolerance via de novo induction of Tr1 cells in the recipients. Acute viral infection with lymphocytic choriomeningitis virus (LCMV) had no impact on Tr1 cell number and function, neither on the Tr1 cells infused nor on the ones induced, and that was reflected in the robust maintenance of the graft. Moreover, Tr1 cell immunotherapy had no detrimental effect on CD8 and CD4 anti-LCMV effector T-cell responses and viral control. Together, these data suggest that Tr1 cells did not convert to effector cells during acute infection with LCMV, maintained transplant tolerance and did not inhibit antiviral immunity.


Assuntos
Tolerância Imunológica/imunologia , Imunoterapia Adotiva , Transplante das Ilhotas Pancreáticas , Coriomeningite Linfocítica/imunologia , Linfócitos T Reguladores/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Diabetes Mellitus Tipo 1/terapia , Ilhotas Pancreáticas , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Modelos Animais , Linfócitos T Reguladores/transplante
4.
J Autoimmun ; 67: 82-89, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26553386

RESUMO

The induction of innate and adaptive immunity is essential for controlling viral infections. Limited or overwhelming innate immunity can negatively impair the adaptive immune response. Therefore, balancing innate immunity separately from activating the adaptive immune response would result in a better antiviral immune response. Recently, we demonstrated that Usp18-dependent replication of virus in secondary lymphatic organs contributes to activation of the innate and adaptive immune responses. Whether specific mechanisms can balance innate and adaptive immunity separately remains unknown. In this study, using lymphocytic choriomeningitis virus (LCMV) and replication-deficient single-cycle LCMV vectors, we found that viral replication of the initial inoculum is essential for activating virus-specific CD8(+) T cells. In contrast, extracellular distribution of virus along the splenic conduits is necessary for inducing systemic levels of type I interferon (IFN-I). Although enforced virus replication is driven primarily by Usp18, B cell-derived lymphotoxin beta contributes to the extracellular distribution of virus along the splenic conduits. Therefore, lymphotoxin beta regulates IFN-I induction independently of CD8(+) T-cell activity. We found that two separate mechanisms act together in the spleen to guarantee amplification of virus during infection, thereby balancing the activation of the innate and adaptive immune system.


Assuntos
Imunidade Adaptativa , Imunidade Inata , Replicação Viral/imunologia , Animais , Modelos Animais de Doenças , Humanos , Interferon Tipo I/biossíntese , Linfonodos/imunologia , Linfonodos/metabolismo , Linfonodos/virologia , Ativação Linfocitária , Linfócitos/imunologia , Linfócitos/metabolismo , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/fisiologia , Linfotoxina-beta/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Baço/imunologia , Baço/metabolismo , Baço/virologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Ubiquitina Tiolesterase/genética , Ubiquitina Tiolesterase/metabolismo
5.
Viruses ; 16(3)2024 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-38543756

RESUMO

CD8+ T cells are critical to the adaptive immune response against viral pathogens. However, overwhelming antigen exposure can result in their exhaustion, characterised by reduced effector function, failure to clear virus, and the upregulation of inhibitory receptors, including programmed cell death 1 (PD-1). However, exhausted T cell responses can be "re-invigorated" by inhibiting PD-1 or the primary ligand of PD-1: PD-L1. Further, the absence of the type I interferon receptor IFNAR1 also results in T cell exhaustion and virus persistence in lymphocytic choriomeningitis virus Armstrong (LCMV-Arm)-infected mice. In this study, utilizing single- and double-knockout mice, we aimed to determine whether ablation of PD-1 could restore T cell functionality in the absence of IFNAR1 signalling in LCMV-Arm-infected mice. Surprisingly, this did not re-invigorate the T cell response and instead, it converted chronic LCMV-Arm infection into a lethal disease characterized by severe lung inflammation with an infiltration of neutrophils and T cells. Depletion of CD8+ T cells, but not neutrophils, rescued mice from lethal disease, demonstrating that IFNAR1 is required to prevent T cell exhaustion and virus persistence in LCMV-Arm infection, and in the absence of IFNAR1, PD-L1 is required for survival. This reveals an important interplay between IFNAR1 and PD-L1 with implications for therapeutics targeting these pathways.


Assuntos
Interferon Tipo I , Coriomeningite Linfocítica , Camundongos , Animais , Vírus da Coriomeningite Linfocítica , Linfócitos T CD8-Positivos , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/metabolismo , Camundongos Knockout , Interferon Tipo I/metabolismo , Camundongos Endogâmicos C57BL
6.
J Vet Med Sci ; 85(3): 393-398, 2023 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-36754384

RESUMO

Lymphocytic choriomeningitis virus (LCMV) is a neglected rodent-borne zoonotic virus in the genus Mammarenavirus and family Arenaviridae, that can cause aseptic meningitis in humans. A recent study identified infectious LCMV in ticks in northeastern (NE) China. To explore the distribution of LCMV, we determined the prevalence and genetically characterized LCMV in ticks in Jilin Province, NE China. Ticks collected in Huadian, Dunhua, and Jiaohe were pooled and LCMV was detected using reverse transcription polymerase chain reaction. The complete genomes of the LCMV-positive pools were amplified and used for phylogenetic analysis. A total of 1679 questing and engorged ticks were collected and divided into 170 pools, including Ixodes persulcatus (5%), Dermacentor silvarum (89%), and Haemaphysalis japonica (6%). Twenty-four pools of D. silvarum (14.9%, 95% CI:9.5-22.3) and three pools of H. japonica (36.3%, 95% CI:9.8-99.5) collected from cattle were LCMV-positive. No I. persulcatus pools were identified as LCMV-positive. Two complete genome sequences (strains JL-DH01 and JL-DH02) were successfully amplified. They had nucleotide identities of 96.4-99.8% with strains JX31, JX14, DH46, and JX4 identified in ticks from Jilin Province. Phylogenetic analysis indicated that JL-DH01 and JL-DH02 clustered with Jilin strains in the same branch and belonged to genotype I. The findings add to the knowledge of the genetic diversity and geographical distribution of LCMV in ticks in NE China.


Assuntos
Ixodes , Ixodidae , Humanos , Animais , Bovinos , Vírus da Coriomeningite Linfocítica , Filogenia , China/epidemiologia
7.
Front Immunol ; 14: 1221511, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37662932

RESUMO

During recent years, evidence has emerged that immune privileged sites such as the CNS and the retina may be more integrated in the systemic response to infection than was previously believed. In line with this, it was recently shown that a systemic acute virus infection leads to infiltration of CD8 T cells in the brains of immunocompetent mice. In this study, we extend these findings to the neurological tissue of the eye, namely the retina. We show that an acute systemic virus infection in mice leads to a transient CD8 T cell infiltration in the retina that is not directed by virus infection inside the retina. CD8 T cells were found throughout the retinal tissue, and had a high expression of CXCR6 and CXCR3, as also reported for tissue residing CD8 T cells in the lung and liver. We also show that the pigment epithelium lining the retina expresses CXCL16 (the ligand for CXCR6) similar to epithelial cells of the lung. Thus, our results suggest that the retina undergoes immune surveillance during a systemic infection, and that this surveillance appears to be directed by mechanisms similar to those described for non-privileged tissues.


Assuntos
Sepse , Viroses , Animais , Camundongos , Encéfalo , Linfócitos T CD8-Positivos , Quimiocina CXCL16 , Retina
8.
Front Immunol ; 13: 908707, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35958615

RESUMO

T cell-based therapies have been widely explored for the treatment of cancer and chronic infection, but B cell-based therapies have remained largely unexplored. To study the effect of B cell therapy, we adoptively transferred virus-specific B cells into mice that were chronically infected with lymphocytic choriomeningitis virus (LCMV). Adoptive transfer of virus-specific B cells resulted in increase in antibody titers and reduction of viral loads. Importantly, the efficacy of B cell therapy was partly dependent on antibody effector functions, and was improved by co-transferring virus-specific CD4 T cells. These findings provide a proof-of-concept that adoptive B cell therapy can be effective for the treatment of chronic infections, but provision of virus-specific CD4 T cells may be critical for optimal virus neutralization.


Assuntos
Coriomeningite Linfocítica , Animais , Terapia Baseada em Transplante de Células e Tecidos , Imunoterapia Adotiva , Coriomeningite Linfocítica/terapia , Vírus da Coriomeningite Linfocítica , Camundongos , Camundongos Endogâmicos C57BL
9.
Cell Rep ; 38(5): 110303, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35108544

RESUMO

Across the animal kingdom, multivalency discriminates antibodies from all other immunoglobulin superfamily members. The evolutionary forces conserving multivalency above other structural hallmarks of antibodies remain, however, incompletely defined. Here, we engineer monovalent either Fc-competent or -deficient antibody formats to investigate mechanisms of protection of neutralizing antibodies (nAbs) and non-neutralizing antibodies (nnAbs) in virus-infected mice. Antibody bivalency enables the tethering of virions to the infected cell surface, inhibits the release of virions in cell culture, and suppresses viral loads in vivo independently of Fc gamma receptor (FcγR) interactions. In return, monovalent antibody formats either do not inhibit virion release and fail to protect in vivo or their protective efficacy is largely FcγR dependent. Protection in mice correlates with virus-release-inhibiting activity of nAb and nnAb rather than with their neutralizing capacity. These observations provide mechanistic insights into the evolutionary conservation of antibody bivalency and help refining correlates of nnAb protection for vaccine development.


Assuntos
Anticorpos Antivirais/farmacologia , Antivirais/farmacologia , Anticorpos Anti-HIV/farmacologia , Receptores Fc/efeitos dos fármacos , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/farmacologia , Anticorpos Antivirais/imunologia , Epitopos/efeitos dos fármacos , Epitopos/imunologia , Anticorpos Anti-HIV/imunologia , Imunoglobulina G/efeitos dos fármacos , Imunoglobulina G/imunologia , Camundongos Endogâmicos C57BL , Receptores de IgG/efeitos dos fármacos , Receptores de IgG/imunologia
10.
Antiviral Res ; 197: 105230, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34965446

RESUMO

Lassa virus (LASV) belongs to the Old World genus Mammarenavirus, family Arenaviridae, and order Bunyavirales. Arenavirus contains a segmented negative-sense RNA genome, which is in line with the bunyavirus and orthomyxoviruses. The segmented negative-sense RNA viruses utilize a cap-snatching strategy to provide primers cleavaged from the host capped mRNA for viral mRNA transcription. As a similar strategy and the conformational conservation shared with these viruses, the endonuclease (EN) would serve as an attractive target for developing broad-spectrum inhibitors. Using the LASV minigenome (MG) system, we screened a fragment-based drug discovery library and found that two hits, F1204 and F1781, inhibited LASV MG activity. Both hits also inhibited the prototype arenavirus Lymphocytic choriomeningitis virus (LCMV) MG activity. Furthermore, both hits effectively inhibited authentic LCMV and severe fever with thrombocytopenia syndrome virus (SFTSV) infections. Similarly, both hits could inhibit the activity of LASV, LCMV, and SFTSV EN. The combination of either compound with an arenavirus entry inhibitor had significant synergistic antiviral effects. Moreover, both hits were found to be capable of binding to LASV EN with a binding affinity at the micromolar level. These findings provide a basis for developing the hits as potential candidates for the treatment of segmented negative-sense RNA virus infections.


Assuntos
Antivirais/farmacologia , Descoberta de Drogas/métodos , Endonucleases/antagonistas & inibidores , Vírus Lassa/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Internalização do Vírus/efeitos dos fármacos , Animais , Antivirais/isolamento & purificação , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Células HEK293 , Ensaios de Triagem em Larga Escala/métodos , Humanos , Febre Lassa/tratamento farmacológico , Vírus Lassa/enzimologia , Células Vero
11.
Methods Mol Biol ; 2524: 223-233, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35821475

RESUMO

Reverse genetics systems provide a powerful tool to generate recombinant arenavirus expressing reporters to facilitate the investigation of the arenavirus life cycle and also for the discovery of antiviral countermeasures. The plasmid-encoded viral ribonucleoprotein components initiate the transcription and replication of a plasmid-driven full-length viral genome, resulting in infectious virus. Thereby, this approach is ideal for the generation of recombinant arenaviruses expressing reporter genes that can be used as valid surrogates for virus replication. By splitting the small viral segment (S) into two viral segments (S1 and S2), each of them encoding a reporter gene, recombinant tri-segmented arenavirus can be rescued. Bi-reporter-expressing recombinant tri-segmented arenaviruses represent an excellent tool to study the biology of arenaviruses, including the identification and characterization of both prophylactic and therapeutic countermeasures for the treatment of arenaviral infections. In this chapter, we describe a detailed protocol on the generation and in vitro characterization of recombinant arenaviruses containing a tri-segment genome expressing two reporter genes based on the prototype member in the family, lymphocytic choriomeningitis virus (LCMV). Similar experimental approaches can be used for the generation of bi-reporter-expressing tri-segment recombinant viruses for other members in the arenavirus family.


Assuntos
Infecções por Arenaviridae , Genética Reversa , Infecções por Arenaviridae/genética , Infecções por Arenaviridae/prevenção & controle , Genes Reporter , Humanos , Vírus da Coriomeningite Linfocítica/genética , Genética Reversa/métodos , Replicação Viral/genética
12.
Viruses ; 13(7)2021 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-34202565

RESUMO

Arenaviruses and coronaviruses include several human pathogenic viruses, such as Lassa virus, Lymphocytic choriomeningitis virus (LCMV), SARS-CoV, MERS-CoV, and SARS-CoV-2. Although these viruses belong to different virus families, they possess a common motif, the DED/EDh motif, known as an exonuclease (ExoN) motif. In this study, proof-of-concept studies, in which the DED/EDh motif in these viral proteins, NP for arenaviruses, and nsp14 for coronaviruses, could be a drug target, were performed. Docking simulation studies between two structurally different chemical compounds, ATA and PV6R, and the DED/EDh motifs in these viral proteins indicated that these compounds target DED/EDh motifs. The concentration which exhibited modest cell toxicity was used with these compounds to treat LCMV and SARS-CoV-2 infections in two different cell lines, A549 and Vero 76 cells. Both ATA and PV6R inhibited the post-entry step of LCMV and SARS-CoV-2 infection. These studies strongly suggest that DED/EDh motifs in these viral proteins could be a drug target to combat two distinct viral families, arenaviruses and coronaviruses.


Assuntos
Antivirais/farmacologia , Exorribonucleases/antagonistas & inibidores , Vírus da Coriomeningite Linfocítica/efeitos dos fármacos , SARS-CoV-2/efeitos dos fármacos , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas Virais/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos , Células A549 , Motivos de Aminoácidos , Animais , Chlorocebus aethiops , Descoberta de Drogas , Humanos , Simulação de Acoplamento Molecular , Células Vero
13.
Viruses ; 13(4)2021 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-33919632

RESUMO

Rodent-borne arenaviruses have been traditionally predominantly associated with certain muroid species from Mastomys/Praomys genera (African arenaviruses) or with species that belong to murid subfamily Cricetidae (New World arenaviruses) [...].


Assuntos
Infecções por Arenaviridae/veterinária , Arenavirus/genética , Arenavirus/patogenicidade , Sequência de Aminoácidos , Animais , Infecções por Arenaviridae/transmissão , Arenavirus/classificação , Peixes/virologia , Humanos , Roedores/virologia , Serpentes/virologia
14.
Viruses ; 11(10)2019 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-31546586

RESUMO

Viral infections are a natural part of our existence. They can affect us in many ways that are the result of the interaction between the viral pathogen and our immune system. Most times, the resulting immune response is beneficial for the host. The pathogen is cleared, thus protecting our vital organs with no other consequences. Conversely, the reaction of our immune system against the pathogen can cause organ damage (immunopathology) or lead to autoimmune disease. To date, there are several mechanisms for virus-induced autoimmune disease, including molecular mimicry and bystander activation, in support of the "fertile field" hypothesis (terms defined in our review). In contrast, viral infections have been associated with protection from autoimmunity through mechanisms that include Treg invigoration and immune deviation, in support of the "hygiene hypothesis", also defined here. Infection with lymphocytic choriomeningitis virus (LCMV) is one of the prototypes showing that the interaction of our immune system with viruses can either accelerate or prevent autoimmunity. Studies using mouse models of LCMV have helped conceive and establish several concepts that we now know and use to explain how viruses can lead to autoimmune activation or induce tolerance. Some of the most important mechanisms established during the course of LCMV infection are described in this short review.


Assuntos
Infecções por Arenaviridae/imunologia , Doenças Autoimunes/imunologia , Tolerância Imunológica/imunologia , Vírus da Coriomeningite Linfocítica/fisiologia , Animais , Doenças Autoimunes/virologia , Autoimunidade/imunologia , Humanos , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Modelos Imunológicos
15.
Math Biosci Eng ; 16(6): 7009-7021, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31698601

RESUMO

We fit an immune response model to data reporting the CD4+ T cell numbers from the 28 days following the infection of mice with lymphocytic choriomeningitis virus LCMV. We used an ODE model that was previously used to describe qualitatively the behaviour of CD4+ T cells, regulatory T cells (Tregs) and interleukine-2 (IL-2) density. The model considered two clonotypes of T cells in order to fit simultaneously the two time series for the gp61 and NP309 epitopes. We observed the proliferation of T cells and, to a lower extent, Tregs during the immune activation phase following infection and subsequently, during the contraction phase, a smooth transition from faster to slower death rates. The six parameters that were optimized were: the beginning and ending times of the immune response, the growth rate of T cells, their capacity, and the two related with the homeostatic numbers of T cells that respond to each epitope. We showed that the ODE model was able to be calibrated thus providing a quantitative description of the data.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Sistema Imunitário , Linfócitos/imunologia , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica , Animais , Antígenos/imunologia , Simulação por Computador , Epitopos/imunologia , Homeostase , Coriomeningite Linfocítica/sangue , Camundongos , Modelos Estatísticos , Fenótipo , Linfócitos T/citologia
16.
Viruses ; 10(2)2018 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-29439402

RESUMO

Mammarenavirusesare single-stranded RNA viruses with a bisegmented ambisense genome. Ingestion has been shown as a natural route of transmission for both Lassa virus (LASV) and Lymphocytic choriomeningitis virus (LCMV). Due to the mechanism of transmission, epithelial tissues are among the first host cells to come in contact with the viruses, and as such they potentially play a role in spread of virus to naïve hosts. The role of the intestinal epithelia during arenavirus infection remains to be uncharacterized. We have utilized a well-established cell culture model, Caco-2, to investigate the role of intestinal epithelia during intragastric infection. We found that LCMV-Armstrong, LCMV-WE, and Mopeia (MOPV) release infectious progeny via similar patterns. However, the reassortant virus, ML-29, containing the L segment of MOPV and S segment of LASV, exhibits a unique pattern of viral release relative to LCMV and MOPV. Furthermore, we have determined attachment efficacy to Caco-2 cells is potentially responsible for observed replication kinetics of these viruses in a polarized Caco-2 cell model. Collectively, our data shows that viral dissemination and interaction with intestinal epithelia may be host, tissue, and viral specific.


Assuntos
Arenavirus/fisiologia , Mucosa Intestinal/virologia , Animais , Infecções por Arenaviridae/virologia , Células CACO-2 , Linhagem Celular , Células Cultivadas , Chlorocebus aethiops , Humanos , Vírus Reordenados , Células Vero , Internalização do Vírus , Replicação Viral
18.
J Neuropathol Exp Neurol ; 75(11): 1031-1047, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27667772

RESUMO

Lymphocytic choriomeningitis virus (LCMV) infection during pregnancy injures the human fetal brain. Neonatal rats inoculated with LCMV are an excellent model of congenital LCMV infection because they develop cerebellar injuries similar to those in humans. To evaluate the role of T-lymphocytes in LCMV-induced cerebellar pathology, congenitally athymic rats, deficient in T-lymphocytes were compared with euthymic rats. Peak viral titers and cellular targets of infection were similar, but viral clearance from astrocytes was impaired in the athymic rats. Cytokines and chemokines rose to higher levels and for a greater duration in the euthymic rats than in their athymic counterparts. The euthymic rats developed an intense lymphocytic infiltration, accompanied by destructive lesions of the cerebellum and a neuronal migration defect because of T-cell-mediated alteration of Bergmann glia. These pathologic changes were absent in the athymic rats but were restored by adoptive transfer of lymphocytes. Athymic rats were not free of pathologic effects, however, as the virus induced cerebellar hypoplasia. Thus, T-lymphocytes play key roles in LCMV clearance, cytokine/chemokine responses, and pathogenesis of destructive lesions and neuronal migration disturbances but not all pathology is T-lymphocyte-dependent. Cerebellar hypoplasia from LCMV occurs even in the absence of T-lymphocytes and is likely due to the viral infection itself.

19.
Mol Immunol ; 67(2 Pt B): 625-35, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26319106

RESUMO

The etiology of multiple sclerosis (MS) involves both genetic and environmental factors. Genetically, the strongest link is with HLA DRB1*1501, but the environmental trigger, probably a virus, remains uncertain. This investigation scans a panel of proteins from encephalitogenic viruses for peptides homologous to the primary autoantigen from myelin basic protein (MBP), then evaluates candidate peptides against a motif required for T cell cross-reactivity and compares viral prevalence patterns to epidemiological characteristics of MS. The only peptide meeting criteria for cross-reactivity with MBP was one from lymphocytic choriomeningitis virus (LCMV), a zoonotic agent. In contrast to current candidates such as Epstein-Barr virus, the distribution of LCMV is consistent with epidemiological features of MS, including concentration in the temperate zone, higher prevalence farther from the equator, and increased prevalence in proximity to regions of peak MS incidence, while lack of person-to-person transmission is consistent with low MS concordance across monozygotic twins. Further, LCMV blocks induction of type I interferon (IFN). Hypothetically this would dysregulate immune processes in favor of proinflammatory pathways as well as upregulating HLA class II and providing more binding sites for autoantigen. The combination of molecular mimicry with virally-induced immune dysregulation has the potential to explain aspects of autoimmunity not addressed by either mechanism alone.


Assuntos
Motivos de Aminoácidos , Vírus da Coriomeningite Linfocítica/fisiologia , Esclerose Múltipla/virologia , Peptídeos/química , Sequência de Aminoácidos , Geografia , Alemanha/epidemiologia , Haplótipos/genética , Humanos , Dados de Sequência Molecular , Esclerose Múltipla/epidemiologia , Esclerose Múltipla/imunologia , Ligação Proteica , Homologia de Sequência de Aminoácidos , Linfócitos T/imunologia , Proteínas Virais/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA