Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 414
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Avian Pathol ; 53(4): 303-311, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38411905

RESUMO

Monitoring Marek's disease (MD) vaccination is routinely done by evaluating the load of MD vaccine in the feather pulp (FP) between 7 and 10 days of age. However, attempts in our laboratory to detect a novel CVI-LTR vaccine in the FP samples from commercial flocks failed. The objective of this study was to evaluate the most suitable tissue and age to monitor CVI-LTR vaccination. We used two different commercial CVI988 vaccines as controls. One hundred and sixty 1-day-old commercial brown layers were vaccinated with either CVI-LTR, CVI988-A, CVI988-B or remained unvaccinated. Samples of the spleen, thymus, and bursa were collected at 3, 4, 5, and 6 days of age and samples of FP were collected at 7 and 21 days for DNA isolation. Our results showed that CVI-LTR replicated earlier than CVI988 vaccines in the lymphoid organs but was not detected in the FP at either 7 or at 21 days of age. We also confirmed that either the spleen or thymus collected at 4-6 days was a suitable sample to monitor CVI-LTR vaccination in commercial flocks. Finally, we evaluated the load of oncogenic MDV DNA in five commercial flocks that were vaccinated with either CVI-LTR + rHVT or CVI988-A + rHVT. The load of oncogenic MDV DNA was evaluated at 21 days in the FP in 20 chickens per group. Our results demonstrated that CVI-LTR was more successful in reducing oncogenic MDV DNA at 21 days of age than the CVI988-A strain.RESEARCH HIGHLIGHTSCVI-LTR replicates in the thymus and spleen earlier than CVI988.CVI-LTR replicates in lymphoid organs but it cannot be detected in feather pulp.CVI-LTR reduced the load of oncogenic MDV DNA more efficiently than CVI988.


Assuntos
Galinhas , Plumas , Vacinas contra Doença de Marek , Doença de Marek , Baço , Timo , Animais , Galinhas/virologia , Doença de Marek/prevenção & controle , Doença de Marek/virologia , Vacinas contra Doença de Marek/imunologia , Baço/virologia , Plumas/virologia , Timo/virologia , Doenças das Aves Domésticas/virologia , Doenças das Aves Domésticas/prevenção & controle , Sequências Repetidas Terminais , Feminino , Vacinação/veterinária , Bolsa de Fabricius/virologia , Vírus da Reticuloendoteliose/genética , Herpesvirus Galináceo 2/genética , Replicação Viral , DNA Viral/genética
2.
Proc Natl Acad Sci U S A ; 117(10): 5420-5429, 2020 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-32094187

RESUMO

Chronic infection provokes alterations in inflammatory and suppressive pathways that potentially affect the function and integrity of multiple tissues, impacting both ongoing immune control and restorative immune therapies. Here we demonstrate that chronic lymphocytic choriomeningitis virus infection rapidly triggers severe thymic depletion, mediated by CD8 T cell-intrinsic type I interferon (IFN) and signal transducer and activator of transcription 2 (Stat2) signaling. Occurring temporal to T cell exhaustion, thymic cellularity reconstituted despite ongoing viral replication, with a rapid secondary thymic depletion following immune restoration by anti-programmed death-ligand 1 (PDL1) blockade. Therapeutic hematopoietic stem cell transplant (HSCT) during chronic infection generated new antiviral CD8 T cells, despite sustained virus replication in the thymus, indicating an impairment in negative selection. Consequently, low amounts of high-affinity self-reactive T cells also escaped the thymus following HSCT during chronic infection. Thus, by altering the stringency and partially impairing negative selection, the host generates new virus-specific T cells to replenish the fight against the chronic infection, but also has the potentially dangerous effect of enabling the escape of self-reactive T cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Interferon Tipo I/metabolismo , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/patologia , Vírus da Coriomeningite Linfocítica , Timo/patologia , Timo/virologia , Animais , Atrofia/virologia , Antígeno B7-H1/antagonistas & inibidores , Doença Crônica , Transplante de Células-Tronco Hematopoéticas , Interferon Tipo I/genética , Coriomeningite Linfocítica/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator de Transcrição STAT2/metabolismo , Transdução de Sinais , Replicação Viral
3.
J Virol ; 94(21)2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-32817213

RESUMO

While feline leukemia virus (FeLV) has been shown to infect felid species other than the endemic domestic cat host, differences in FeLV susceptibility among species has not been evaluated. Previous reports have noted a negative correlation between endogenous FeLV (enFeLV) copy number and exogenous FeLV (exFeLV) infection outcomes in domestic cats. Since felids outside the genus Felis do not harbor enFeLV genomes, we hypothesized absence of enFeLV results in more severe disease consequences in felid species lacking these genomic elements. We infected primary fibroblasts isolated from domestic cats (Felis catus) and pumas (Puma concolor) with FeLV and quantitated proviral and viral antigen loads. Domestic cat enFeLV env and long terminal repeat (LTR) copy numbers were determined for each individual and compared to FeLV viral outcomes. FeLV proviral and antigen levels were also measured in 6 naturally infected domestic cats and 11 naturally infected Florida panthers (P. concolor coryi). We demonstrated that puma fibroblasts are more permissive to FeLV than domestic cat cells, and domestic cat FeLV restriction was highly related to enFeLV-LTR copy number. Terminal tissues from FeLV-infected Florida panthers and domestic cats had similar exFeLV proviral copy numbers, but Florida panther tissues have higher FeLV antigen loads. Our work indicates that enFeLV-LTR elements negatively correlate with exogenous FeLV replication. Further, Puma concolor samples lacking enFeLV are more permissive to FeLV infection than domestic cat samples, suggesting that endogenization can play a beneficial role in mitigating exogenous retroviral infections. Conversely, presence of endogenous retroelements may relate to new host susceptibility during viral spillover events.IMPORTANCE Feline leukemia virus (FeLV) can infect a variety of felid species. Only the primary domestic cat host and related small cat species harbor a related endogenous virus in their genomes. Previous studies noted a negative association between the endogenous virus copy number and exogenous virus infection in domestic cats. This report shows that puma cells, which lack endogenous FeLV, produce more virus more rapidly than domestic cat fibroblasts following cell culture challenge. We document a strong association between domestic cat cell susceptibility and FeLV long terminal repeat (LTR) copy number, similar to observations in natural FeLV infections. Viral replication does not, however, correlate with FeLV env copy number, suggesting that this effect is specific to FeLV-LTR elements. This discovery indicates a protective capacity of the endogenous virus against the exogenous form, either via direct interference or indirectly via gene regulation, and may suggest evolutionary outcomes of retroviral endogenization.


Assuntos
Variações do Número de Cópias de DNA , Produtos do Gene env/genética , Vírus da Leucemia Felina/genética , Vírus da Leucemia Felina/patogenicidade , Leucemia Felina/virologia , Puma/virologia , Animais , Medula Óssea/patologia , Medula Óssea/virologia , Gatos , Feminino , Fibroblastos/patologia , Fibroblastos/virologia , Produtos do Gene env/metabolismo , Especificidade de Hospedeiro , Vírus da Leucemia Felina/metabolismo , Leucemia Felina/patologia , Linfonodos/patologia , Linfonodos/virologia , Masculino , Cultura Primária de Células , Baço/patologia , Baço/virologia , Sequências Repetidas Terminais , Timo/patologia , Timo/virologia , Carga Viral , Replicação Viral/genética
4.
Can J Microbiol ; 67(1): 23-28, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32640169

RESUMO

Understanding the pathogenesis of certain viral agents is essential for developing new treatments and obtaining a clinical cure. With the onset of the new coronavirus (SARS-CoV-2) pandemic in the beginning of 2020, a rush to conduct studies and develop drugs has led to the publication of articles that seek to address knowledge gaps and contribute to the global scientific research community. There are still no reports on the infectivity or repercussions of SARS-CoV-2 infection on the central lymphoid organ, the thymus, nor on thymocytes or thymic epithelial cells. In this brief review, we present a hypothesis about lymphopenia observed in SARS patients and the probable pathological changes that the thymus may undergo due to this new virus.


Assuntos
COVID-19/complicações , COVID-19/imunologia , Linfopenia/complicações , Timo/virologia , Animais , Humanos , Linfopenia/imunologia , Linfopenia/virologia , Camundongos , Modelos Imunológicos , Pandemias , Timo/imunologia
5.
Proc Natl Acad Sci U S A ; 115(45): 11603-11607, 2018 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-30337483

RESUMO

Marek's disease virus (MDV) is a highly oncogenic alphaherpesvirus that causes immunosuppression, paralysis, and deadly lymphomas in chickens. In infected animals, B cells are efficiently infected and are thought to amplify the virus and transfer it to T cells. MDV subsequently establishes latency in T cells and transforms CD4+ T cells, resulting in fatal lymphomas. Despite many years of research, the exact role of the different B and T cell subsets in MDV pathogenesis remains poorly understood, mostly due to the lack of reverse genetics in chickens. Recently, Ig heavy chain J gene segment knockout (JH-KO) chickens lacking mature and peripheral B cells have been generated. To determine the role of these B cells in MDV pathogenesis, we infected JH-KO chickens with the very virulent MDV RB1B strain. Surprisingly, viral load in the blood of infected animals was not altered in the absence of B cells. More importantly, disease and tumor incidence in JH-KO chickens was comparable to wild-type animals, suggesting that both mature and peripheral B cells are dispensable for MDV pathogenesis. Intriguingly, MDV efficiently replicated in the bursa of Fabricius in JH-KO animals, while spread of the virus to the spleen and thymus was delayed. In the absence of B cells, MDV readily infected CD4+ and CD8+ T cells, allowing efficient virus replication in the lymphoid organs and transformation of T cells. Taken together, our data change the dogma of the central role of B cells, and thereby provide important insights into MDV pathogenesis.


Assuntos
Linfócitos B/imunologia , Genoma Viral , Herpesvirus Galináceo 2/patogenicidade , Linfoma/patologia , Doença de Marek/patologia , Vírus Oncogênicos/patogenicidade , Animais , Animais Geneticamente Modificados , Animais Recém-Nascidos , Bolsa de Fabricius/imunologia , Bolsa de Fabricius/virologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Embrião de Galinha , Galinhas , DNA Viral/genética , DNA Viral/imunologia , Herpesvirus Galináceo 2/genética , Herpesvirus Galináceo 2/imunologia , Cadeias Pesadas de Imunoglobulinas/genética , Contagem de Linfócitos , Linfoma/genética , Linfoma/imunologia , Linfoma/virologia , Doença de Marek/genética , Doença de Marek/imunologia , Doença de Marek/virologia , Vírus Oncogênicos/genética , Vírus Oncogênicos/imunologia , Baço/imunologia , Baço/virologia , Timo/imunologia , Timo/virologia , Carga Viral , Virulência , Replicação Viral
6.
J Infect Dis ; 218(suppl_5): S409-S417, 2018 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-30085162

RESUMO

Ebola virus (EBOV) and Marburg virus (MARV) outbreaks are highly lethal, and infection results in a hemorrhagic fever with complex etiology. These zoonotic viruses dysregulate the immune system to cause disease, in part by replicating within myeloid cells that would normally innately control viral infection and shape the adaptive immune response. We used triple knockout (TKO)-bone marrow, liver, thymus (BLT) humanized mice to recapitulate the early in vivo human immune response to filovirus infection. Disease severity in TKO-BLT mice was dissimilar between EBOV and MARV with greater severity observed during EBOV infection. Disease severity was related to increased Kupffer cell infection in the liver, higher levels of myeloid dysfunction, and skewing of macrophage subtypes in EBOV compared with MARV-infected mice. Overall, the TKO-BLT model provided a practical in vivo platform to study the human immune response to filovirus infection and generated a better understanding of how these viruses modulate specific components of the immune system.


Assuntos
Medula Óssea/virologia , Ebolavirus/patogenicidade , Marburgvirus/patogenicidade , Células Mieloides/virologia , Timo/virologia , Animais , Medula Óssea/imunologia , Ebolavirus/imunologia , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/virologia , Imunidade/imunologia , Fígado/imunologia , Fígado/virologia , Macrófagos/imunologia , Macrófagos/virologia , Doença do Vírus de Marburg/imunologia , Doença do Vírus de Marburg/virologia , Marburgvirus/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/imunologia , Timo/imunologia , Virulência/imunologia
7.
BMC Genomics ; 19(1): 211, 2018 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-29558886

RESUMO

BACKGROUND: Ranaviruses (family Iridoviridae, nucleocytoplasmic large DNA viruses) have been reported as promiscuous pathogens of cold-blooded vertebrates. Rana grylio virus (RGV, a ranavirus), from diseased frog Rana grylio with a genome of 105.79 kb and Andrias davidianus ranavirus (ADRV), from diseased Chinese giant salamander (CGS) with a genome of 106.73 kb, contains 99% homologous genes. RESULTS: To uncover the differences in virus replication and host responses under interspecies infection, we analyzed transcriptomes of CGS challenged with RGV and ADRV in different time points (1d, 7d) for the first time. A total of 128,533 unigenes were obtained from 820,858,128 clean reads. Transcriptome analysis revealed stronger gene expression of RGV than ADRV at 1 d post infection (dpi), which was supported by infection in vitro. RGV replicated faster and had higher titers than ADRV in cultured CGS cell line. RT-qPCR revealed the RGV genes including the immediate early gene (RGV-89R) had higher expression level than that of ADRV at 1 dpi. It further verified the acute infection of RGV in interspecies infection. The number of differentially expressed genes and enriched pathways from RGV were lower than that from ADRV, which reflected the variant host responses at transcriptional level. No obvious changes of key components in pathway "Antigen processing and presentation" were detected for RGV at 1 dpi. Contrarily, ADRV infection down-regulated the expression levels of MHC I and CD8. The divergent host immune responses revealed the differences between interspecies and natural infection, which may resulted in different fates of the two viruses. Altogether, these results revealed the differences in transcriptome responses among ranavirus interspecies infection of amphibian and new insights in DNA virus-host interactions in interspecies infection. CONCLUSION: The DNA virus (RGV) not only expressed self-genes and replicated quickly after entry into host under interspecies infection, but also avoided the over-activation of host responses. The strategy could gain time for the survival of interspecies pathogen, and may provide opportunity for its adaptive evolution and interspecies transmission.


Assuntos
Infecções por Vírus de DNA/veterinária , Interações Hospedeiro-Patógeno , Ranavirus/genética , Ranidae , Análise de Sequência de DNA/veterinária , Urodelos , Animais , Infecções por Vírus de DNA/virologia , Genoma Viral , Sequenciamento de Nucleotídeos em Larga Escala , Ranidae/genética , Ranidae/virologia , Timo/virologia , Transcriptoma , Urodelos/genética , Urodelos/virologia , Proteínas Virais/genética , Replicação Viral
8.
Biochem Biophys Res Commun ; 502(4): 472-478, 2018 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-29856997

RESUMO

Both the lung and the thymus are vital target organ for pathogens including viruses. The immunoproteasome (i-proteasome) enhances antigen presentation for MHC class I molecules to activate CD8+T lymphocyte. These facilitate antiviral adaptive immune response. Our previous study found that, expression of i-proteasome subunits in porcine lung was altered during normal and inflammatory conditions. To date, the expression of i-proteasome subunits in porcine thymus to viruses has not been investigated. In the present study, LMP2, LMP7, and MECL-1 were cloned, identified and their sequences encoded predicted proteins of 216, 275, and 278 amino acids, respectively. Expression of LMP2, LMP7, and MECL-1, in the cytoplasm and nucleus, was markedly altered in the porcine reproductive and respiratory syndrome virus (PRRSV)-infected lung and thymus. And dendritic cells and epithelial cells readily expressed the i-proteasome subunit LMP2 in the thymus of PRRSV-infected pigs compared to that in mock-infected pigs. Additionally, the in vitro stimulation of a PAM cell line with PolyI:C for 12 and 24 h resulted in increased LMP2, LMP7, and MECL-1 expression. These results suggest a central role for these complexes in the activation of an antiviral immune response in pigs. A better understanding of the role of the i-proteasome in different cell types, tissues, and hosts could improve vaccine design and facilitate the development of effective treatment strategies for viral infections.


Assuntos
Pulmão/imunologia , Complexo de Endopeptidases do Proteassoma/imunologia , Suínos/imunologia , Timo/imunologia , Sequência de Aminoácidos , Animais , Apresentação de Antígeno , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Cisteína Endopeptidases/química , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/imunologia , Pulmão/virologia , Filogenia , Síndrome Respiratória e Reprodutiva Suína/imunologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/patogenicidade , Complexo de Endopeptidases do Proteassoma/química , Complexo de Endopeptidases do Proteassoma/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Homologia de Sequência de Aminoácidos , Suínos/genética , Suínos/virologia , Timo/virologia
9.
J Virol ; 91(9)2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28179532

RESUMO

The human roseoloviruses human herpesvirus 6A (HHV-6A), HHV-6B, and HHV-7 comprise the Roseolovirus genus of the human Betaherpesvirinae subfamily. Infections with these viruses have been implicated in many diseases; however, it has been challenging to establish infections with roseoloviruses as direct drivers of pathology, because they are nearly ubiquitous and display species-specific tropism. Furthermore, controlled study of infection has been hampered by the lack of experimental models, and until now, a mouse roseolovirus has not been identified. Herein we describe a virus that causes severe thymic necrosis in neonatal mice, characterized by a loss of CD4+ T cells. These phenotypes resemble those caused by the previously described mouse thymic virus (MTV), a putative herpesvirus that has not been molecularly characterized. By next-generation sequencing of infected tissue homogenates, we assembled a contiguous 174-kb genome sequence containing 128 unique predicted open reading frames (ORFs), many of which were most closely related to herpesvirus genes. Moreover, the structure of the virus genome and phylogenetic analysis of multiple genes strongly suggested that this virus is a betaherpesvirus more closely related to the roseoloviruses, HHV-6A, HHV-6B, and HHV-7, than to another murine betaherpesvirus, mouse cytomegalovirus (MCMV). As such, we have named this virus murine roseolovirus (MRV) because these data strongly suggest that MRV is a mouse homolog of HHV-6A, HHV-6B, and HHV-7.IMPORTANCE Herein we describe the complete genome sequence of a novel murine herpesvirus. By sequence and phylogenetic analyses, we show that it is a betaherpesvirus most closely related to the roseoloviruses, human herpesviruses 6A, 6B, and 7. These data combined with physiological similarities with human roseoloviruses collectively suggest that this virus is a murine roseolovirus (MRV), the first definitively described rodent roseolovirus, to our knowledge. Many biological and clinical ramifications of roseolovirus infection in humans have been hypothesized, but studies showing definitive causative relationships between infection and disease susceptibility are lacking. Here we show that MRV infects the thymus and causes T-cell depletion, suggesting that other roseoloviruses may have similar properties.


Assuntos
Modelos Animais de Doenças , Herpesviridae/classificação , Herpesvirus Humano 6/genética , Herpesvirus Humano 7/genética , Depleção Linfocítica , Infecções por Roseolovirus/virologia , Animais , Sequência de Bases , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , DNA Viral/genética , Genoma Viral/genética , Humanos , Evasão da Resposta Imune/genética , Evasão da Resposta Imune/imunologia , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Fases de Leitura Aberta/genética , Filogenia , Análise de Sequência de DNA , Timo/virologia
10.
Clin Exp Immunol ; 187(3): 399-407, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27790717

RESUMO

Type B coxsackievirus (CV-B) infections are involved frequently in the triggering of several autoimmune diseases such as myocarditis, dilated cardiomyopathy, pericarditis, pancreatitis, type 1 diabetes, encephalitis, thyroiditis or Sjögren's syndrome. Serological and virological evidence suggests that maternal infections during pregnancy can play a role in the appearance of these diseases in offspring. The current study aims to explore the effect of an in-utero CV-B infection on the fetal thymus, the central site for programming immunological self-tolerance. In this perspective, female Swiss albino mice were inoculated intraperitoneally or orally with the diabetogenic CV-B4 E2 strain at gestational days 10 or 17. Offspring were killed at different post-inoculation times, and their thymuses were analysed for evidence of infection and alterations in thymic T cell subsets. In-utero CV-B infection of the thymus was demonstrated during the course of vertical transmission, as attested by viral RNA and infectious virus detection in most analysed samples. No histopathological changes were evident. Thymic T cells were not depleted, despite being positive for viral RNA. As evidenced by flow cytometry analysis, CV-B infection of the fetal thymus induced significant changes of thymic T cell populations, particularly with maternal inoculation at gestational day 10. Altogether, these findings suggest that CV-B infection of the fetal thymus may play an important role in the genesis of autoimmune diseases.


Assuntos
Doenças Autoimunes/virologia , Infecções por Coxsackievirus/virologia , Enterovirus Humano B/imunologia , Timo/virologia , Útero/virologia , Animais , Doenças Autoimunes/imunologia , Infecções por Coxsackievirus/imunologia , Feminino , Tolerância Imunológica/imunologia , Transmissão Vertical de Doenças Infecciosas , Masculino , Camundongos , Gravidez , RNA Viral/genética , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/virologia , Timo/imunologia , Útero/imunologia
11.
J Neurovirol ; 23(4): 520-538, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28321697

RESUMO

Varicella zoster virus (VZV) causes varicella during acute infection and establishes latency in the sensory ganglia. Reactivation of VZV results in herpes zoster, a debilitating and painful disease. It is believed that VZV reactivates due to a decline in cell-mediated immunity; however, the roles that CD4 versus CD8 T cells play in the prevention of herpes zoster remain poorly understood. To address this question, we used a well-characterized model of VZV infection where rhesus macaques are intrabronchially infected with the homologous simian varicella virus (SVV). Latently infected rhesus macaques were thymectomized and depleted of either CD4 or CD8 T cells to induce selective senescence of each T cell subset. After T cell depletion, the animals were transferred to a new housing room to induce stress. SVV reactivation (viremia in the absence of rash) was detected in three out of six CD8-depleted and two out of six CD4-depleted animals suggesting that both CD4 and CD8 T cells play a critical role in preventing SVV reactivation. Viral loads in multiple ganglia were higher in reactivated animals compared to non-reactivated animals. In addition, reactivation results in sustained transcriptional changes in the ganglia that enriched to gene ontology and diseases terms associated with neuronal function and inflammation indicative of potential damage as a result of viral reactivation. These studies support the critical role of cellular immunity in preventing varicella virus reactivation and indicate that reactivation results in long-lasting remodeling of the ganglia transcriptome.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Gânglios Sensitivos/imunologia , Herpes Zoster/veterinária , Herpesvirus Humano 3/imunologia , Proteínas do Tecido Nervoso/genética , Ativação Viral/imunologia , Animais , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/virologia , Feminino , Gânglios Sensitivos/virologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Ontologia Genética , Herpes Zoster/genética , Herpes Zoster/imunologia , Depleção Linfocítica/métodos , Macaca mulatta , Masculino , Anotação de Sequência Molecular , Proteínas do Tecido Nervoso/imunologia , Estresse Psicológico , Timectomia , Timo/imunologia , Timo/cirurgia , Timo/virologia
12.
Arch Virol ; 162(11): 3473-3480, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28795249

RESUMO

Naïve pregnant cattle exposed to pestiviruses between 40-125 days of gestation can give birth to persistently infected (PI) calves. Clinical presentation and survivability, in PI cattle, is highly variable even with the same pestivirus strain whereas the clinical presentation in acute infections is more uniform with severity of symptoms being primarily a function of virulence of the infecting virus. The aim of this study was to compare thymic depletion, as measured by comparing the area of the thymic cortex to the medulla (corticomedullary ratio), in acute and persistent infections of the same pestivirus isolate. The same general trends were observed with each pestivirus isolate. Thymic depletion was observed in both acutely and persistently infected calves. The average thymic depletion observed in acutely infected calves was greater than that in age matched PI calves. PI calves, regardless of infecting virus, revealed a greater variability in amount of depletion compared to acutely infected calves. A trend was observed between survivability and depletion of the thymus, with PI calves surviving less than 5 weeks having lower corticomedullary ratios and greater depletion. This is the first study to compare PI and acutely infected calves with the same isolates as well as to evaluate PI calves based on survivability. Further, this study identified a quantifiable phenotype associated with potential survivability.


Assuntos
Vírus da Diarreia Viral Bovina Tipo 1 , Vírus da Diarreia Viral Bovina Tipo 2 , Linfócitos/patologia , Infecções por Pestivirus/veterinária , Pestivirus/classificação , Timo/citologia , Animais , Bovinos , Vírus da Diarreia Viral Bovina Tipo 1/patogenicidade , Vírus da Diarreia Viral Bovina Tipo 2/patogenicidade , Pestivirus/patogenicidade , Infecções por Pestivirus/patologia , Infecções por Pestivirus/virologia , Timo/patologia , Timo/virologia , Virulência
13.
BMC Infect Dis ; 17(1): 18, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-28056841

RESUMO

BACKGROUND: Respiratory syncytial virus (RSV) infection is an important cause of hospitalization in previously healthy infants. Immunological mechanisms predisposing infants to severe disease are poorly understood. Early biomarkers for disease severity may assist clinical decisions. We investigated T-cell receptor excision circles (TREC), episomal DNA made during thymic T-cell receptor rearrangement, and a marker for thymus activity, both during disease and in neonatal screening cards as a risk factor for RSV disease severity. METHODS: One hundred thirteen patients hospitalized with RSV infection <12 months of age, grouped by disease severity, were available for this investigation, in which we conducted both a prospective and a case-control study. The prospective study included 47 RSV positive infants (mild n = 13, moderate n = 10, severe n = 24). TREC counts were determined by PCR of DNA extracted from EDTA-blood collected on hospitalization, and corrected for lymphocytes using ANCOVA. The case-control study included 85 newborns who later in infancy became RSV positive (mild n = 32, moderate n = 24, severe n = 29) and 47 newborns who never developed RSV disease as healthy controls included from health centres in the same catchment area. TRECs were measured using DNA extracted from dry blood spots from stored neonatal screening cards, followed by PCR. Student's T-test compared patients with controls, ANOVA compared disease severity groups. RESULTS: During RSV infection patients in the severe disease group had significantly lower (p = 0.017) TREC/200 µL blood compared to the other two disease groups, after correction for lymphocyte count. Newborn TREC levels, were significantly higher in RSV patients compared to controls (p < 0.0001). No significant differences in TREC copies at birth were found between disease severities. CONCLUSION: During acute RSV infection a lower number of TREC is found in the severe disease group. TREC has potential as an immunological marker for severe RSV infection. Higher neonatal TREC counts indicate that infants later presenting with severe RSV do not have reduced thymic activity at birth and probably no congenital T-cell defect.


Assuntos
DNA/sangue , Receptores de Antígenos de Linfócitos T/genética , Infecções por Vírus Respiratório Sincicial/genética , Timo/virologia , Estudos de Casos e Controles , Teste em Amostras de Sangue Seco , Feminino , Humanos , Lactente , Recém-Nascido , Contagem de Linfócitos , Masculino , Estudos Prospectivos , Infecções por Vírus Respiratório Sincicial/etiologia , Infecções por Vírus Respiratório Sincicial/imunologia
14.
J Immunol ; 194(5): 2260-7, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25609844

RESUMO

The study of T cell immunity at barrier surfaces has largely focused on T cells bearing the αß TCR. However, T cells that express the γδ TCR are disproportionately represented in peripheral tissues of mice and humans, suggesting they too may play an important role responding to external stimuli. In this article, we report that, in a murine model of cutaneous infection with vaccinia virus, dermal γδ T cell numbers increased 10-fold in the infected ear and resulted in a novel γδ T cell population not found in naive skin. Circulating γδ T cells were specifically recruited to the site of inflammation and differentially contributed to dermal populations based on their CD27 expression. Recruited γδ T cells, the majority of which were CD27(+), were granzyme B(+) and made up about half of the dermal population at the peak of the response. In contrast, recruited and resident γδ T cell populations that made IL-17 were CD27(-). Using a double-chimera model that can discriminate between the resident dermal and recruited γδ T cell populations, we demonstrated their divergent functions and contributions to early stages of tissue inflammation. Specifically, the loss of the perinatal thymus-derived resident dermal population resulted in decreased cellularity and collateral damage in the tissue during viral infection. These findings have important implications for our understanding of immune coordination at barrier surfaces and the contribution of innate-like lymphocytes on the front lines of immune defense.


Assuntos
Derme/imunologia , Orelha/virologia , Infecções por Poxviridae/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Subpopulações de Linfócitos T/imunologia , Vaccinia virus/imunologia , Animais , Movimento Celular , Quimera/imunologia , Quimera/virologia , Derme/patologia , Derme/virologia , Orelha/patologia , Regulação da Expressão Gênica , Granzimas/genética , Granzimas/imunologia , Imunidade Inata , Interleucina-17/genética , Interleucina-17/imunologia , Linfonodos/imunologia , Linfonodos/patologia , Linfonodos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Poxviridae/patologia , Infecções por Poxviridae/virologia , Receptores de Antígenos de Linfócitos T gama-delta/genética , Transdução de Sinais , Baço/imunologia , Baço/patologia , Baço/virologia , Subpopulações de Linfócitos T/patologia , Subpopulações de Linfócitos T/virologia , Timo/imunologia , Timo/patologia , Timo/virologia , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia
15.
J Virol ; 89(4): 2201-8, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25473058

RESUMO

UNLABELLED: A unique HIV-host equilibrium exists in untreated HIV-2-infected individuals. This equilibrium is characterized by low to undetectable levels of viremia throughout the disease course, despite the establishment of disseminated HIV-2 reservoirs at levels comparable to those observed in untreated HIV-1 infection. Although the clinical spectrum is similar in the two infections, HIV-2 infection is associated with a much lower rate of CD4 T-cell decline and has a limited impact on the mortality of infected adults. Here we investigated HIV-2 infection of the human thymus, the primary organ for T-cell production. Human thymic tissue and suspensions of total or purified CD4 single-positive thymocytes were infected with HIV-2 or HIV-1 primary isolates using either CCR5 or CXCR4 coreceptors. We found that HIV-2 infected both thymic organ cultures and thymocyte suspensions, as attested to by the total HIV DNA and cell-associated viral mRNA levels. Nevertheless, thymocytes featured reduced levels of intracellular Gag viral protein, irrespective of HIV-2 coreceptor tropism and cell differentiation stage, in agreement with the low viral load in culture supernatants. Our data show that HIV-2 is able to infect the human thymus, but the HIV-2 replication cycle in thymocytes is impaired, providing a new model to identify therapeutic targets for viral replication control. IMPORTANCE: HIV-1 infects the thymus, leading to a decrease in CD4 T-cell production that contributes to the characteristic CD4 T-cell loss. HIV-2 infection is associated with a very low rate of progression to AIDS and is therefore considered a unique naturally occurring model of attenuated HIV disease. HIV-2-infected individuals feature low to undetectable plasma viral loads, in spite of the numbers of circulating infected T cells being similar to those found in patients infected with HIV-1. We assessed, for the first time, the direct impact of HIV-2 infection on the human thymus. We show that HIV-2 is able to infect the thymus but that the HIV-2 replication cycle in thymocytes is impaired. We propose that this system will be important to devise immunotherapies that target viral production, aiding the design of future therapeutic strategies for HIV control.


Assuntos
HIV-2/fisiologia , Interações Hospedeiro-Patógeno , Timócitos/virologia , Timo/virologia , Replicação Viral , Adulto , Células Cultivadas , Pré-Escolar , HIV-1/fisiologia , Humanos , Lactente , Recém-Nascido , Técnicas de Cultura de Órgãos , Timo/patologia
16.
PLoS Pathog ; 10(3): e1003937, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24651250

RESUMO

In chronic viral infections, persistent antigen presentation causes progressive exhaustion of virus-specific CD8+ T cells. It has become clear, however, that virus-specific naïve CD8+ T cells newly generated from the thymus can be primed with persisting antigens. In the setting of low antigen density and resolved inflammation, newly primed CD8+ T cells are preferentially recruited into the functional memory pool. Thus, continual recruitment of naïve CD8+ T cells from the thymus is important for preserving the population of functional memory CD8+ T cells in chronically infected animals. Friend virus (FV) is the pathogenic murine retrovirus that establishes chronic infection in adult mice, which is bolstered by the profound exhaustion of virus-specific CD8+ T cells induced during the early phase of infection. Here we show an additional evasion strategy in which FV disseminates efficiently into the thymus, ultimately leading to clonal deletion of thymocytes that are reactive to FV antigens. Owing to the resultant lack of virus-specific recent thymic emigrants, along with the above exhaustion of antigen-experienced peripheral CD8+ T cells, mice chronically infected with FV fail to establish a functional virus-specific CD8+ T cell pool, and are highly susceptible to challenge with tumor cells expressing FV-encoded antigen. However, FV-specific naïve CD8+ T cells generated in uninfected mice can be primed and differentiate into functional memory CD8+ T cells upon their transfer into chronically infected animals. These findings indicate that virus-induced central tolerance that develops during the chronic phase of infection accelerates the accumulation of dysfunctional memory CD8+ T cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Tolerância Imunológica/imunologia , Memória Imunológica/imunologia , Infecções por Retroviridae/imunologia , Timo/virologia , Envelhecimento , Animais , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular/imunologia , Doença Crônica , Feminino , Citometria de Fluxo , Vírus da Leucemia Murina de Friend/imunologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos , Timo/imunologia
17.
Virol J ; 13(1): 199, 2016 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-27903277

RESUMO

BACKGROUND: We previously reported that a clinical isolate of dengue virus (DENV) is capable of causing acute-phase systemic infection in mice harboring knockouts of the genes encoding type-I and -II interferon IFN receptors (IFN-α/ß/γR KO mice); in contrast, other virulent DENV isolates exhibited slow disease progression in this mice, yielding lethal infection around 20 days post-infection (p.i.). In the present study, we sought to clarify the dynamics of slow disease progression by examining disease progression of a type-2 DENV clinical isolate (DV2P04/08) in mice. METHODS: The tissue distributions of DV2P04/08 in several organs of infeted mice were examined at different time points. Whole genome viral sequences from organs were determined. RESULTS: At day 6 p.i., high levels of viral RNA (vRNA) were detected in non-neuronal organs (including peritoneal exudate cells (PECs), spleen, kidney, liver, lung, and bone marrow) but not in brain. By day 14 p.i, vRNA levels subsequently decreased in most organs, with the exception of thymus and brain. Sequence analysis of the whole genome of the original P04/08 and those of viruses recovered from mouse brain and thymus demonstrated the presence of both synonymous and non-synonymous mutations. Individual mice showed different virus populations in the brain. The vRNA sequence derived from brain of one mouse was nearly identical to the original DV2P04/08 inoculum, suggesting that there was no need for adaptation of DV2P04/08 for growth in the brain. However, quasispecies (that is, mixed populations, detected as apparent nucleotide mixtures during sequencing) were observed in the thymus of another mouse, and interestingly only mutant population invaded the brain at a late stage of infection. CONCLUSIONS: These results suggested that the mouse nearly succeeded in eliminating virus from non-neuronal organs but failed to do so from brain. Although the cause of death by DV2P04/08 infection is likely to be the result of virus invasion to brain, its processes to the death are different in individual mice. This study will provide a new insight into disease progression of DENV in mice.


Assuntos
Encéfalo/virologia , Vírus da Dengue/classificação , Vírus da Dengue/genética , Dengue/virologia , Variação Genética , Receptores de Interferon/deficiência , Timo/virologia , Animais , Vírus da Dengue/isolamento & purificação , Modelos Animais de Doenças , Feminino , Genoma Viral , Masculino , Camundongos , Camundongos Knockout , Análise de Sequência de DNA , Análise de Sobrevida , Virulência
18.
Vet Res ; 47(1): 76, 2016 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-27494990

RESUMO

Several routes of porcine reproductive and respiratory virus PRRSV transmission across the porcine diffuse epitheliochorial placentation have been proposed, but none have been proven. The objectives of this study were to investigate associations between numbers of CD163 and CD169 positive macrophages, cathepsin positive areolae, and type 2 PRRSV load at the maternal-fetal interface in order to examine important factors related to transplacental infection. On gestation day 85 ± 1, naïve pregnant gilts were inoculated with PRRSV (n = 114) or were sham inoculated (n = 19). At 21 days post-inoculation (dpi), dams and their litters were humanely euthanized and necropsied. Samples of the maternal-fetal interface (uterus with fully attached placenta) and fetal thymus were collected for analysis by RT-qPCR to quantify PRRSV RNA concentration. The corresponding paraffin-embedded uterine tissue sections were subjected to immunohistochemistry for PRRSV nucleocapsid N protein, CD163, CD169, and cathepsin. Our findings confirm significant increases in the numbers of PRRSV, CD163 and CD169 positive cells at the maternal-fetal interface during type 2 PRRSV infection in pregnant gilts. PRRSV load in fetal thymus was positively related to CD163(+) cell count in endometrium and negatively related to CD163(+) cell count in placenta, but unrelated to CD169 counts or cathepsin positive areolae. The endometrium:placenta ratio of CD163 cells, and to a lesser extent CD169 cells, was significantly associated with an increase fetal viral load in thymus. These findings suggest a more important role for CD163(+) cells following trans-placental PRRSV infection, but dichotomous responses in endometrium and placenta for both CD163 and CD169 cells.


Assuntos
Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/imunologia , Endométrio/virologia , Macrófagos/imunologia , Placenta/virologia , Síndrome Respiratória e Reprodutiva Suína/imunologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/fisiologia , Receptores de Superfície Celular/imunologia , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Timo/virologia , Animais , Contagem de Células/veterinária , Endométrio/imunologia , Feminino , Placenta/imunologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Gravidez , Suínos , Timo/embriologia , Timo/imunologia , Carga Viral
19.
Genet Mol Res ; 15(3)2016 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-27706718

RESUMO

DNA methylation is an important epigenetic modification in eukaryotes, which plays a significant role in regulating gene expression. When the host is invaded by the influenza virus, gene expression is regulated via changes in DNA methylation levels or patterns, leading to the activation or suppression of relevant signaling pathways or networks, triggering a series of immune responses against viral invasion. Here, we investigated the changes in genomic DNA methylation in the immune organs of chicken infected with H5N1 influenza virus. Genome-wide DNA methylation levels in the spleen, thymus, and bursa of Fabricius of specific pathogen-free (SPF) chicken infected with the Guangdong (G-H5N1) and Anhui (A-H5N1) H5N1 strains, and water (control) were analyzed by fluorescence-labeled methylation-sensitive amplified polymorphism (F-MSAP). The results indicated that total DNA methylation levels did not differ between spleen genomic DNA in chicken treated with different viral strains and the control (P > 0.05). However, the total DNA methylation levels were significantly upregulated in the thymus (P < 0.01) and bursa (P < 0.05) of chicken in the A-H5N1 group compared to those in the G-H5N1 and control groups. These results provide a basis for the screening of avian influenza-resistance genes or methylation markers, analyzing the epigenetic regulation mechanisms of avian influenza, and performing selective breeding for disease resistance.


Assuntos
Metilação de DNA/genética , Resistência à Doença/genética , Virus da Influenza A Subtipo H5N1/patogenicidade , Influenza Aviária/genética , Animais , Bolsa de Fabricius/imunologia , Bolsa de Fabricius/virologia , Galinhas , Metilação de DNA/imunologia , Resistência à Doença/imunologia , Epigênese Genética , Genoma/genética , Virus da Influenza A Subtipo H5N1/imunologia , Influenza Aviária/imunologia , Influenza Aviária/virologia , Transdução de Sinais , Baço/imunologia , Baço/virologia , Timo/imunologia , Timo/virologia
20.
Retrovirology ; 12: 93, 2015 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-26559632

RESUMO

BACKGROUND: Determining the anatomic compartments that contribute to plasma HIV-1 is critical to understanding the sources of residual viremia during combination antiretroviral therapy (ART). We analyzed viral DNA and RNA populations in the plasma and tissues from macaques infected with SIV containing HIV-1 RT (RT-SHIV) to identify possible sources of persistent viremia and to investigate the effect of ART on viral replication in tissues. Tissues were collected at necropsy from four pigtailed macaques infected for 30 weeks with a diverse population of RT-SHIV. Two animals (6760 and 8232) were untreated and two animals (8030 and 8272) were treated with efavirenz, tenofovir, and emtricitabine for 20 weeks. RESULTS: A total of 1800 single-genome RT-SHIV pol and env DNA and RNA sequences were analyzed from the plasma, PBMCs, axillary and mesenteric lymph nodes, spleen, thymus, small intestine, bone marrow, lung, and brain. Analyses of intracellular DNA and RNA populations revealed that the majority of proviruses in tissues from untreated animal 8232 were not expressed, whereas a greater proportion of proviruses in tissues were expressed from 6760. Few intracellular RNA sequences were detected in treated animals and most contained inactivating mutations, such as frame shifts or large deletions. Phylogenetics showed that RT-SHIV DNA populations in tissues were not different from virus in contemporary plasma samples in the treated or untreated animals, demonstrating a lack of anatomic compartmentalization and suggesting that plasma viremia is derived from multiple tissue sources. No sequence divergence was detected in the plasma or between tissues in the treated animals after 20 weeks of ART indicating a lack of ongoing replication in tissues during treatment. CONCLUSIONS: Virus populations in plasma and tissues did not differ significantly in either treated or untreated macaques, suggesting frequent exchange of virus or infected cells between tissues and plasma, consistent with non-compartmentalized and widely disseminated infection. There was no genetic evidence of ongoing replication in tissues during suppressive ART.


Assuntos
Fármacos Anti-HIV/uso terapêutico , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/fisiologia , Replicação Viral/efeitos dos fármacos , Animais , Fármacos Anti-HIV/administração & dosagem , Terapia Antirretroviral de Alta Atividade , Medula Óssea/virologia , Encéfalo/virologia , DNA Viral/sangue , Modelos Animais de Doenças , Farmacorresistência Viral , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , Pulmão/virologia , Linfonodos/virologia , Macaca mulatta , Filogenia , RNA Viral/sangue , Vírus da Imunodeficiência Símia/genética , Baço/virologia , Timo/virologia , Carga Viral/efeitos dos fármacos , Viremia/tratamento farmacológico , Viremia/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA