Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Eur J Med Res ; 27(1): 5, 2022 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-35022077

RESUMO

BACKGROUND: Intervertebral disc degeneration (IDD) is a natural progression of age-related processes. Associated with IDD, degenerative disc disease (DDD) is a pathologic condition implicated as a major cause of chronic lower back pain, which can have a severe impact on the quality of life of patients. As degeneration progression is associated with elevated levels of inflammatory cytokines, enhanced aggrecan and collagen degradation, and changes in the disc cell phenotype. The purpose of this study was to investigate the biological and cytological characteristics of rabbit nucleus pulposus mesenchymal stem cells (NPMSCs)-a key factor in IDD-and to determine the effect of the growth and differentiation factor-5 (GDF5) on the differentiation of rabbit NPMSCs transduced with a lentivirus vector. METHODS: An in vitro culture model of rabbit NPMSCs was established and NPMSCs were identified by flow cytometry (FCM) and quantitative real-time PCR (qRT-PCR). Subsequently, NPMSCs were randomly divided into three groups: a transfection group (the lentiviral vector carrying GDF5 gene used to transfect NPMSCs); a control virus group (the NPMSCs transfected with an ordinary lentiviral vector); and a normal group (the NPMSCs alone). FCM, qRT-PCR, and western blot (WB) were used to detect the changes in NPMSCs. RESULTS: The GDF5-transfected NPMSCs displayed an elongated shape, with decreased cell density, and significantly increased GDF5 positivity rate in the transfected group compared to the other two groups (P < 0.01). The mRNA levels of Krt8, Krt18, and Krt19 in the transfected group were significantly higher in comparison with the other two groups (P < 0.01), and the WB results were consistent with that of qRT-PCR. CONCLUSIONS: GDF5 could induce the differentiation of NPMSCs. The lentiviral vector carrying the GDF5 gene could be integrated into the chromosome genome of NPMSCs and promoted differentiation of NPMSCs into nucleus pulposus cells. Our findings advance the development of feasible and effective therapies for IDD.


Assuntos
Regulação da Expressão Gênica , Fator 5 de Diferenciação de Crescimento/genética , Infecções por Lentivirus/virologia , Lentivirus , Células-Tronco Mesenquimais/citologia , Núcleo Pulposo/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Fator 5 de Diferenciação de Crescimento/biossíntese , Infecções por Lentivirus/metabolismo , Infecções por Lentivirus/patologia , Células-Tronco Mesenquimais/virologia , Núcleo Pulposo/patologia , Núcleo Pulposo/virologia , Coelhos
2.
J Virol ; 95(13): e0017821, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-33762419

RESUMO

As the hosts of lentiviruses, almost 40 species of felids (family Felidae) are distributed around the world, and more than 20 feline species test positive for feline immunodeficiency virus (FIV), a lineage of lentiviruses. These observations suggest that FIVs globally infected a variety of feline species through multiple cross-species transmission events during a million-year history. Cellular restriction factors potentially inhibit lentiviral replication and limit cross-species lentiviral transmission, and cellular APOBEC3 deaminases are known as a potent restriction factor. In contrast, lentiviruses have evolutionary-acquired viral infectivity factor (Vif) to neutralize the APOBEC3-mediated antiviral effect. Because the APOBEC3-Vif interaction is strictly specific for viruses and their hosts, a comprehensive investigation focusing on Vif-APOBEC3 interplay can provide clues that will elucidate the roles of this virus-host interplay on cross-species transmission of lentiviruses. Here, we performed a comprehensive investigation with 144 patterns of a round robin test using 18 feline APOBEC3Z3 genes, an antiviral APOBEC3 gene in felid, and 8 FIV Vifs and derived a matrix showing the interplay between feline APOBEC3Z3 and FIV Vif. We particularly focused on the interplay between the APOBEC3Z3 of three felids (domestic cat, ocelot, and Asian golden cat) and an FIV Vif (strain Petaluma), and revealed that residues 65 and 66 of the APOBEC3Z3 protein of multiple felids are responsible for the counteraction triggered by FIV Petaluma Vif. Altogether, our findings can be a clue to elucidate not only the scenarios of the cross-species transmissions of FIVs in felids but also the evolutionary interaction between mammals and lentiviruses. IMPORTANCE Most of the emergences of new virus infections originate from the cross-species transmission of viruses. The fact that some virus infections are strictly specific for the host species indicates that certain "species barriers" in the hosts restrict cross-species jump of viruses, while viruses have evolutionary acquired their own "arms" to overcome/antagonize/neutralize these hurdles. Therefore, understanding of the molecular mechanism leading to successful cross-species viral transmission is crucial for considering the menus of the emergence of novel pathogenic viruses. In the field of retrovirology, APOBEC3-Vif interaction is a well-studied example of the battles between hosts and viruses. Here, we determined the sequences of 11 novel feline APOBEC3Z3 genes and demonstrated that all 18 different feline APOBEC3Z3 proteins tested exhibit anti-feline immunodeficiency virus (FIV) activity. Our comprehensive investigation focusing on the interplay between feline APOBEC3 and FIV Vif can be a clue to elucidate the scenarios of the cross-species transmissions of FIVs in felids.


Assuntos
Desaminase APOBEC-1/metabolismo , Produtos do Gene vif/metabolismo , Vírus da Imunodeficiência Felina/metabolismo , Infecções por Lentivirus/transmissão , Animais , Gatos , Linhagem Celular , Células HEK293 , Especificidade de Hospedeiro/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Infecções por Lentivirus/patologia , Panthera , Replicação Viral/fisiologia
3.
Viruses ; 10(6)2018 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-29890760

RESUMO

Small Ruminant Lentiviruses (SRLVs) are widespread in many countries and cause economically relevant, slow, and persistent diseases in sheep and goats. Monitoring the genetic diversity of SRLVs is useful to improve the diagnostic tools used in the eradication programs. In this study, SRLVs detected in Spanish Assaf sheep with different grades of lymphoproliferative mastitis were sequenced. Genetic characterization showed that most samples belonged to type A and were closer to Spanish SRLV isolates previously classified as A2/A3. Four samples belonged to subtype B2 and showed higher homology with Italian B2 strains than with Spanish B2 isolates. Amino acid sequences of immuno-dominant epitopes in the gag region were very conserved while more alterations were found in the LTR sequences. No significant correlations were found between grades of mastitis and alterations in the sequences although samples with similar histological features were phylogenetically closer to each other. Broader genetic characterization surveys in samples with different grades of SRLV-lesions are required for evaluating potential correlations between SRLV sequences and the severity of diseases.


Assuntos
Variação Genética , Infecções por Lentivirus/veterinária , Lentivirus/classificação , Lentivirus/isolamento & purificação , Glândulas Mamárias Animais/patologia , Doenças dos Ovinos/virologia , Animais , Genótipo , Lentivirus/genética , Infecções por Lentivirus/patologia , Infecções por Lentivirus/virologia , Filogenia , Análise de Sequência de DNA , Homologia de Sequência , Ovinos , Doenças dos Ovinos/patologia , Espanha
4.
Vet Pathol ; 54(3): 413-424, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28113037

RESUMO

The objective of this work was to comparatively study the tissue tropism and the associated pathology of 2 autochthonous small ruminant lentivirus (SRLV) field strains using an experimental infection in sheep through the bone marrow. Fifteen male, SRLV-free lambs of the Rasa Aragonesa breed were inoculated with strain 697 (nervous tissue origin, animals A1-A6), with strain 496 (articular origin, animals B1-B6), or with uninfected culture medium (C1-C3). Clinical, serologic, and polymerase chain reaction (PCR) evaluations were performed periodically. Two lambs from each infected group and a control animal were euthanized at 134, 273, and 319 days postinfection. Tissues were analyzed by gross and histopathologic evaluation; immunohistochemistry for CD3, CD4, CD8, CD68, and FoxP3 cell markers; lung morphometric evaluation; and tissue proviral quantification by PCR. All infected animals became positive either by enzyme-linked immunosorbent assay and/or PCR, with group B lambs showing the highest serologic values and more consistently positive PCR reactions. Group A lambs showed representative lung lesions but only mild histopathologic changes in the central nervous system (CNS) or in carpal joints. Contrarily, group B lambs demonstrated intense carpal arthritis and interstitial pneumonia but an absence of lesions in the CNS. Proviral copies in tissues were detected only in group B lambs. Experimental infection with these SRLV strains indicates that strain 496 is more virulent than strain 697 and more prone to induce arthritis, whereas strain 697 is more likely to reproduce encephalitis in Rasa Aragonesa lambs. Host factors as well as viral factors are responsible for the final clinicopathologic picture during SRLV infections.


Assuntos
Medula Óssea/virologia , Infecções por Lentivirus/veterinária , Lentivirus Ovinos-Caprinos/patogenicidade , Tropismo Viral , Animais , Medula Óssea/patologia , Sistema Nervoso Central/patologia , Sistema Nervoso Central/virologia , Ensaio de Imunoadsorção Enzimática/veterinária , Articulações/patologia , Articulações/virologia , Infecções por Lentivirus/patologia , Infecções por Lentivirus/virologia , Pulmão/patologia , Pulmão/virologia , Masculino , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Ovinos/virologia , Tropismo Viral/fisiologia
5.
J Comp Pathol ; 154(1): 3-8, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26797583

RESUMO

Viral infections have been implicated as the cause of cardiomyopathy in several mammalian species. This study describes hypertrophic cardiomyopathy (HCM) and myocarditis associated with feline immunodeficiency virus (FIV) infection in five cats aged between 1 and 4 years. Clinical manifestations included dyspnoea in four animals, one of which also exhibited restlessness. One animal showed only lethargy, anorexia and vomiting. Necropsy examination revealed marked cardiomegaly, marked left ventricular hypertrophy and pallor of the myocardium and epicardium in all animals. Microscopical and immunohistochemical examination showed multifocal infiltration of the myocardium with T lymphocytes and fewer macrophages, neutrophils and plasma cells. An intense immunoreaction for FIV antigen in the cytoplasm and nucleus of lymphocytes and the cytoplasm of some macrophages was observed via immunohistochemistry (IHC). IHC did not reveal the presence of antigen from feline calicivirus, coronavirus, feline leukaemia virus, feline parvovirus, Chlamydia spp. or Toxoplasma gondii. The results demonstrate the occurrence of FIV infection in inflammatory cells in the myocardium of five cats with myocarditis and HCM.


Assuntos
Cardiomiopatia Hipertrófica/veterinária , Doenças do Gato/virologia , Vírus da Imunodeficiência Felina , Infecções por Lentivirus/veterinária , Miocardite/veterinária , Animais , Cardiomiopatia Hipertrófica/virologia , Gatos , Feminino , Infecções por Lentivirus/patologia , Masculino , Miocardite/virologia
6.
AIDS Res Hum Retroviruses ; 30(1): 58-66, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23924068

RESUMO

Using the feline immunodeficiency virus (FIV) model for AIDS lentivirus infection, we previously demonstrated that Treg cells from FIV-infected cats up-regulate membrane-associated tumor growth factor beta (mTGF-ß) during the course of infection and that activated T lymphocytes up-regulate TGF-ß receptor II (TGF-ßRII) during the course of infection. Furthermore, we have demonstrated that autologous coculture of Tregs with Th cells from FIV-infected cats leads to suppression of interleukin (IL)-2 production and loss of proliferation in a TGF-ß-dependent fashion. Nuclear factor of activated T cells (NFAT) 2 has been identified as integral to effector Th cell maturation and function by promoting IL-2 transcription. Therefore, we questioned whether NFAT2 expression might be altered by TGF-ß signaling. Feline NFAT2 exon sequences were identified based upon sequence homology to human and murine NFAT2. Following stimulation, IL-2 and NFAT2 mRNA levels were similarly increased in both FIV(-) and FIV(+) cats. Activated CD4(+)CD25(-) cells from both FIV(-) and FIV(+) cats cocultured with autologous CD4(+)CD25(+) cells or treated with TGF-ß demonstrated decreased IL-2 production; however, NFAT2 mRNA levels were unaffected. Although NFAT2 mRNA levels were unaffected, chromatin immunoprecipitation (ChIP) for NFAT2 indicated decreased NFAT2 binding at the IL-2 promoter in suppressed Th cells. These data suggest that TGF-ß-mediated Treg cell suppression of IL-2 transcription is modulated through alterations in NFAT2 binding to the IL-2 promoter.


Assuntos
Vírus da Imunodeficiência Felina/imunologia , Interleucina-2/genética , Fatores de Transcrição NFATC/genética , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Reguladores/imunologia , Sequência de Aminoácidos , Animais , Gatos , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Interleucina-2/biossíntese , Infecções por Lentivirus/imunologia , Infecções por Lentivirus/patologia , Infecções por Lentivirus/virologia , Linfonodos/citologia , Linfonodos/virologia , Ativação Linfocitária/imunologia , Dados de Sequência Molecular , Fatores de Transcrição NFATC/antagonistas & inibidores , Regiões Promotoras Genéticas/imunologia , Ligação Proteica/imunologia , RNA Mensageiro/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Alinhamento de Sequência , Linfócitos T Reguladores/virologia , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/farmacologia , Regulação para Cima
7.
J Vet Intern Med ; 27(4): 798-805, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23734699

RESUMO

BACKGROUND: The clinical course and outcome of natural feline immunodeficiency virus (FIV) infection are variable and incompletely understood. Assigning clinical relevance to FIV infection in individual cats represents a considerable clinical challenge. OBJECTIVE: To compare signalment, hematologic and biochemical data, major clinical problem, and survival among client-owned, FIV-infected, and uninfected domestic cats. ANIMALS: Client-owned, domestic cats tested for FIV (n = 520). METHODS: Retrospective, case control study. Logistic regression analyses were conducted to identify risk factors for FIV infection and to compare hematologic and biochemical data between cases and controls, after adjusting for potential confounders. Survival times were compared using Kaplan-Meier curves. RESULTS: The prevalence of FIV infection was 14.6%. Mixed breed, male sex, and older age were risk factors for FIV infection. Hematologic abnormalities, biochemical abnormalities or both were common in both FIV-infected and uninfected cats. Lymphoid malignancies were slightly more common in FIV-infected than uninfected cats. Survival of FIV-infected cats was not significantly different from that of uninfected cats. CONCLUSIONS AND CLINICAL IMPORTANCE: Multiple hematologic and biochemical abnormalities are common in old, sick cats regardless of their FIV status. Their presence should not be assumed to indicate clinical progression of FIV infection. A negative effect of FIV on survival was not apparent in this study.


Assuntos
Doenças do Gato/virologia , Vírus da Imunodeficiência Felina/isolamento & purificação , Infecções por Lentivirus/veterinária , Envelhecimento , Animais , Estudos de Casos e Controles , Doenças do Gato/mortalidade , Doenças do Gato/patologia , Gatos , Feminino , Infecções por Lentivirus/mortalidade , Infecções por Lentivirus/patologia , Vírus da Leucemia Felina/isolamento & purificação , Masculino , Infecções por Retroviridae/veterinária , Infecções por Retroviridae/virologia , Fatores de Risco , Infecções Tumorais por Vírus/veterinária , Infecções Tumorais por Vírus/virologia
8.
PLoS One ; 8(2): e57343, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23460840

RESUMO

SerpinB2, also known as plasminogen activator inhibitor type 2, is a major product of activated monocytes/macrophages and is often strongly induced during infection and inflammation; however, its physiological function remains somewhat elusive. Herein we show that SerpinB2 is induced in peripheral blood mononuclear cells following infection of pigtail macaques with CCR5-utilizing (macrophage-tropic) SIVmac239, but not the rapidly pathogenic CXCR4-utilizing (T cell-tropic) SHIVmn229. To investigate the role of SerpinB2 in lentiviral infections, SerpinB2(-/-) mice were infected with EcoHIV, a chimeric HIV in which HIV gp120 has been replaced with gp80 from ecotropic murine leukemia virus. EcoHIV infected SerpinB2(-/-) mice produced significantly lower anti-gag IgG1 antibody titres than infected SerpinB2(+/+) mice, and showed slightly delayed clearance of EcoHIV. Analyses of published microarray studies showed significantly higher levels of SerpinB2 mRNA in monocytes from HIV-1 infected patients when compared with uninfected controls, as well as a significant negative correlation between SerpinB2 and T-bet mRNA levels in peripheral blood mononuclear cells. These data illustrate that SerpinB2 can be induced by lentiviral infection in vivo and support the emerging notion that a physiological role of SerpinB2 is modulation of Th1/Th2 responses.


Assuntos
Infecções por Lentivirus/imunologia , Inibidor 2 de Ativador de Plasminogênio/metabolismo , Células Th1/imunologia , Células Th2/imunologia , Animais , Movimento Celular , Regulação da Expressão Gênica , Infecções por HIV/genética , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/fisiologia , Humanos , Imunoglobulina G/imunologia , Infecções por Lentivirus/genética , Infecções por Lentivirus/patologia , Infecções por Lentivirus/virologia , Macaca nemestrina/imunologia , Macaca nemestrina/virologia , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Inibidor 2 de Ativador de Plasminogênio/deficiência , Inibidor 2 de Ativador de Plasminogênio/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Vírus da Imunodeficiência Símia/fisiologia , Replicação Viral/fisiologia
9.
J Immunol ; 188(5): 2437-44, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22287715

RESUMO

The molecular mechanisms of acute lung injury are incompletely understood. MicroRNAs (miRNAs) are crucial biological regulators that act by suppressing their target genes and are involved in a variety of pathophysiologic processes. miR-127 appears to be downregulated during lung injury. We set out to investigate the role of miR-127 in lung injury and inflammation. Expression of miR-127 significantly reduced cytokine release by macrophages. Looking into the mechanisms of regulation of inflammation by miR-127, we found that IgG FcγRI (CD64) was a target of miR-127, as evidenced by reduced CD64 protein expression in macrophages overexpressing miR-127. Furthermore, miR-127 significantly reduced the luciferase activity with a reporter construct containing the native 3' untranslated region of CD64. Importantly, we demonstrated that miR-127 attenuated lung inflammation in an IgG immune complex model in vivo. Collectively, these data show that miR-127 targets macrophage CD64 expression and promotes the reduction of lung inflammation. Understanding how miRNAs regulate lung inflammation may represent an attractive way to control inflammation induced by infectious or noninfectious lung injury.


Assuntos
Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/prevenção & controle , Marcação de Genes/métodos , Mediadores da Inflamação/fisiologia , MicroRNAs/fisiologia , Receptores de IgG/metabolismo , Lesão Pulmonar Aguda/patologia , Animais , Complexo Antígeno-Anticorpo/toxicidade , Linhagem Celular , Citocinas/antagonistas & inibidores , Citocinas/metabolismo , Feminino , Células HEK293 , Humanos , Mediadores da Inflamação/metabolismo , Infecções por Lentivirus/imunologia , Infecções por Lentivirus/patologia , Infecções por Lentivirus/prevenção & controle , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/biossíntese , MicroRNAs/genética , Dados de Sequência Molecular , Pneumonia Viral/imunologia , Pneumonia Viral/patologia , Pneumonia Viral/prevenção & controle , Receptores de IgG/antagonistas & inibidores , Receptores de IgG/biossíntese , Receptores de IgG/genética , Células U937
10.
J Immunol ; 184(3): 1566-74, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20042580

RESUMO

Lentivirus infections including HIV and feline immunodeficiency virus (FIV) cause neurovirulence, which is largely mediated by innate immunity. To investigate the interactions between neurovirulence and repeated conditioning by innate immune activation, models of lentivirus infection were exposed to LPS. Gene expression in HIV-infected (HIV+) and control (HIV-) patient brains was compared by real time RT-PCR and immunocytochemistry. Supernatants from mock and HIV-infected monocyte-derived macrophages exposed to LPS were applied to human neurons. FIV-infected (FIV+) and control (FIV-) animals were exposed repeatedly to LPS postinfection together with concurrent neurobehavioral testing, viral load, and host gene analyses. Brains from HIV+ individuals exhibited induction of CD3epsilon, CXCL10, and granzyme A expression (p < 0.05). Supernatants from HIV+ monocyte-derived macrophages induced CXCL10 expression in neurons, which was diminished by IL-10 treatment (p < 0.05). LPS-exposed FIV+ animals demonstrated lower plasma and brain viral loads (p < 0.05). Neuronal CXCL10 expression was increased in FIV+ animals but was suppressed by LPS exposure, together with reduced brain CD3epsilon and granzyme A expression (p < 0.05). In conjunction with preserved NeuN-positive neuronal counts in parietal cortex (p < 0.05), FIV+ animals exposed to LPS also showed less severe neurobehavioral deficits (p < 0.05). Repeated LPS exposures suppressed CXCL10 in the brain and ensuing T cell infiltration with a concomitant reduction in neurovirulence. Thus, innate immune chronic conditioning exerted beneficial effects on neurovirulence through suppression of a specific chemotactic factor, CXCL10, mediated by IL-10, leading to reduced leukocyte infiltration and release of neurotoxic factors.


Assuntos
Encéfalo/imunologia , Quimiocina CXCL10/antagonistas & inibidores , Infecções por HIV/imunologia , Vírus da Imunodeficiência Felina/imunologia , Vírus da Imunodeficiência Felina/patogenicidade , Interleucina-10/fisiologia , Lipopolissacarídeos/farmacologia , Fármacos Neuroprotetores/farmacologia , Animais , Encéfalo/patologia , Encéfalo/virologia , Gatos , Linhagem Celular Tumoral , Movimento Celular/imunologia , Células Cultivadas , Quimiocina CXCL10/fisiologia , Infecções por HIV/patologia , HIV-1/imunologia , Humanos , Vírus da Imunodeficiência Felina/crescimento & desenvolvimento , Infecções por Lentivirus/imunologia , Infecções por Lentivirus/patologia , Infecções por Lentivirus/prevenção & controle , Neurônios/imunologia , Neurônios/patologia , Neurônios/virologia , Carga Viral/imunologia , Virulência/imunologia
11.
Vet Pathol ; 47(1): 148-62, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20080496

RESUMO

Ovine pulmonary adenocarcinoma (OPA) is a naturally occurring and experimentally inducible lung cancer of sheep caused by Jaagsiekte sheep retrovirus (JSRV). The first aim of this study was to monitor the development of OPA with minimally invasive, real-time observations of animals experimentally infected with JSRV as well as ovine lentivirus (maedi-visna virus). Worldwide, simultaneous infection of sheep with these 2 retroviruses is a common occurrence, naturally and experimentally; consequently, the lung tumor homogenates used as inocula contained both viruses. Following inoculation, computed tomography was used to detect tumor nodules early, before the onset of clinical signs, and to monitor tumor advancement. However, not only was OPA disease progression observed, but the apparent spontaneous regression of OPA was witnessed. In fact, regression was more common than progression following JSRV inoculation of neonatal lambs. Immune responses were detected, particularly involving CD3(+) T cells and the production of antibodies against JSRV that may mediate the spontaneous regression of JSRV-induced OPA. The second aim of this study was to determine whether OPA tumors harbor genetic alterations similar to those found in human lung adenocarcinoma. No mutations were found in the tyrosine kinase domain of the epidermal growth factor receptor, KRAS codons 12 and 13, or the DNA-binding domain of p53 in tumor DNA from naturally occurring and experimentally-induced OPA cases. Overall, the genetic profile combined with the disease development data provides further important characterization of OPA and describes, for the first time, spontaneous regression of OPA tumors in experimentally infected sheep.


Assuntos
Retrovirus Jaagsiekte de Ovinos , Infecções por Lentivirus/veterinária , Lentivirus Ovinos-Caprinos , Neoplasias Pulmonares/veterinária , Adenomatose Pulmonar Ovina/patologia , Doenças dos Ovinos/virologia , Animais , DNA Viral/genética , Feminino , Imunidade Humoral , Retrovirus Jaagsiekte de Ovinos/genética , Infecções por Lentivirus/patologia , Infecções por Lentivirus/virologia , Lentivirus Ovinos-Caprinos/genética , Pulmão/patologia , Pulmão/virologia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/virologia , Linfócitos/patologia , Regressão Neoplásica Espontânea/patologia , Testes de Neutralização , Reação em Cadeia da Polimerase/veterinária , Adenomatose Pulmonar Ovina/virologia , Ovinos/virologia , Doenças dos Ovinos/patologia , Tomografia Computadorizada por Raios X
12.
Virology ; 390(1): 1-12, 2009 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-19464039

RESUMO

Feline immunodeficiency virus (FIV) causes AIDS in the domestic cat (Felis catus) but has not been explicitly associated with AIDS pathology in any of the eight free-ranging species of Felidae that are endemic with circulating FIV strains. African lion (Panthera leo) populations are infected with lion-specific FIV strains (FIVple), yet there remains uncertainty about the degree to which FIV infection impacts their health. Reported CD4+ T-lymphocyte depletion in FIVple-infected lions and anecdotal reports of lion morbidity associated with FIV seroprevalence emphasize the concern as to whether FIVple is innocuous or pathogenic. Here we monitored clinical, biochemical, histological and serological parameters among FIVple-positive (N=47) as compared to FIVple-negative (N=17) lions anesthetized and sampled on multiple occasions between 1999 and 2006 in Botswana. Relative to uninfected lions, FIVple-infected lions displayed a significant elevation in the prevalence of AIDS-defining conditions: lymphadenopathy, gingivitis, tongue papillomas, dehydration, and poor coat condition, as well as displaying abnormal red blood cell parameters, depressed serum albumin, and elevated liver enzymes and gamma globulin. Spleen and lymph node biopsies from free-ranging FIVple-infected lions (N=9) revealed evidence of lymphoid depletion, the hallmark pathology documented in immunodeficiency virus infections of humans (HIV-1), macaques, and domestic cats. We conclude that over time FIVple infections in free-ranging lions can lead to adverse clinical, immunological, and pathological outcomes in some individuals that parallel sequelae caused by lentivirus infection in humans (HIV), Asian macaques (SIV) and domestic cats (FIVfca).


Assuntos
Vírus da Imunodeficiência Felina/patogenicidade , Infecções por Lentivirus/veterinária , Leões/virologia , Animais , Animais Selvagens/virologia , Botsuana/epidemiologia , Feminino , Gengivite/patologia , Gengivite/veterinária , Infecções por Lentivirus/epidemiologia , Infecções por Lentivirus/imunologia , Infecções por Lentivirus/patologia , Tecido Linfoide/patologia , Masculino , Neoplasias Bucais/patologia , Neoplasias Bucais/veterinária , Papiloma/patologia , Papiloma/veterinária , Estudos Soroepidemiológicos , Especificidade da Espécie
13.
J Neurosci ; 29(9): 2948-60, 2009 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-19261890

RESUMO

Classical dopaminergic signaling paradigms and emerging studies on direct physical interactions between the D(1) dopamine (DA) receptor and the NMDA glutamate receptor predict a reciprocally facilitating, positive feedback loop. This loop, if not controlled, may cause concomitant overactivation of both D(1) and NMDA receptors, triggering neurotoxicity. Endogenous protective mechanisms must exist. Here, we report that PSD-95, a prototypical structural and signaling scaffold in the postsynaptic density, inhibits D(1)-NMDA receptor subunit 1 (NR1) NMDA receptor association and uncouples NMDA receptor-dependent enhancement of D(1) signaling. This uncoupling is achieved, at least in part, via a disinhibition mechanism by which PSD-95 abolishes NMDA receptor-dependent inhibition of D(1) internalization. Knockdown of PSD-95 immobilizes D(1) receptors on the cell surface and escalates NMDA receptor-dependent D(1) cAMP signaling in neurons. Thus, in addition to its role in receptor stabilization and synaptic plasticity, PSD-95 acts as a brake on the D(1)-NMDA receptor complex and dampens the interaction between them.


Assuntos
Dopamina/fisiologia , Ácido Glutâmico/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas de Membrana/fisiologia , Receptores de Dopamina D1/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Animais , Western Blotting , Linhagem Celular , AMP Cíclico/fisiologia , Proteína 4 Homóloga a Disks-Large , Imunofluorescência , Guanilato Quinases , Humanos , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Infecções por Lentivirus/patologia , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Microscopia Confocal , Plasmídeos , Ensaio Radioligante , Transdução de Sinais/fisiologia , Transfecção
14.
J Neurovirol ; 15(2): 139-52, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19115133

RESUMO

Infection by multiple lentiviral strains is recognized as a major driving force in the human immunodeficiency virus/acquired immunodeficiency syndrome (HIV/AIDS) epidemic, but the neuropathogenic consequences of multivirus infections remain uncertain. Herein, we investigated the neurovirulence and underlying mechanisms of dual lentivirus infections with distinct viral strains. Experimental feline immunodeficiency virus (FIV) infections were performed using cultured cells and an in vivo model of AIDS neuropathogenesis. Dual infections were comprised of two FIV strains (FIV-Ch and FIV-PPR) as copassaged or superinfected viruses, with subsequent outcome analyses of host immune responses, viral load, neuropathological features, and neurobehavioral performance. Dual infections of feline macrophages resulted in greater IL-1beta (interleukin-1beta), TNF-alpha (tumor necrosis factor alpha), and IDO (indoleamine 2,3-dioxygenase) expression and associated neurotoxic properties. FIV coinfection and sequential superinfection in vivo also induced greater IL-1beta, TNF-alpha, and IDO expression in the basal ganglia (BG) and cortex (CTX), compared to the monovirus- and mock-infected groups, although viral loads were similar in single virus- and dual virus-infected animals. Immunoblot analyses disclosed lower synaptophysin immunoreactivity in the CTX resulting from FIV super- and coinfections. Cholinergic and GABAergic neuronal injury was evident in the CTX of animals with dual FIV infections. With increased glial activation and neuronal loss in dual FIV-infected brains, immunohistochemical analysis also revealed elevated detection of cleaved caspase-3 in dysmorphic neurons, which was associated with worsened neurobehavioral abnormalities among animals infected with the copassaged viruses. Dual lentivirus infections caused an escalation in neuroinflammation and ensuing neurodegeneration, underscoring the contribution of infection by multiple viruses to neuropathogenesis.


Assuntos
Vírus da Imunodeficiência Felina/patogenicidade , Inflamação/virologia , Infecções por Lentivirus/patologia , Infecções por Lentivirus/virologia , Degeneração Neural/virologia , Animais , Gatos , Técnicas de Cultura de Células , Cérebro/metabolismo , Feminino , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/biossíntese , Inflamação/imunologia , Inflamação/metabolismo , Interleucina-1beta/biossíntese , Infecções por Lentivirus/imunologia , Infecções por Lentivirus/metabolismo , Contagem de Linfócitos , Degeneração Neural/imunologia , Degeneração Neural/metabolismo , Gravidez , Sinaptofisina/biossíntese , Fator de Necrose Tumoral alfa/biossíntese , Virulência
15.
J Vis Exp ; (14)2008 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-19066578

RESUMO

Pluripotency can be induced in differentiated murine by viral transduction of Oct4, Sox2, Klf4, and c-Myc (Takahashi and Yamanaka, 2006; Wernig, et al., 2007; Okita, et al., 2007; Maherali, et al., 2007). We have devised a reprogramming strategy in which these four transcription factors are expressed from doxycycline (dox)-inducible lentiviral vectors (Brambrink et al., 2008). Using these inducible constructs, we can derive induced pluripotent stem (iPS) cells from mouse embryonic fibroblasts (MEFs). In this video, we demonstrate the procedure for the generation of inducible lentiviruses that express the four transcription factors and show how to infect MEFs with these viruses in order to produce iPS cells. By using inducible lentiviruses, the expression of the four factors in controlled by the addition of doxycyline to the culture medium. The advantage of this system over the traditional retroviral infection is the ability to turn the genes on and off so that the kinetics of reprogramming and gene expression requirements can be analyzed in detail.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Fatores de Transcrição Kruppel-Like/genética , Fator 3 de Transcrição de Octâmero/genética , RNA/genética , Fatores de Transcrição SOXB1/genética , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Animais , Proteínas de Ligação a DNA/biossíntese , Modelos Animais de Doenças , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibroblastos/virologia , Genes myc , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/biossíntese , Infecções por Lentivirus/genética , Infecções por Lentivirus/patologia , Infecções por Lentivirus/virologia , Camundongos , Fator 3 de Transcrição de Octâmero/biossíntese , Fatores de Transcrição SOXB1/biossíntese , alfa-Amilases Salivares , Células-Tronco/citologia , Fatores de Transcrição/biossíntese , Dedos de Zinco
16.
J Immunol ; 179(7): 4732-40, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17878372

RESUMO

HIV-specific CD8+ T cells that secrete multiple cytokines in response to Ag stimulation are associated with the control of virus replication during chronic HIV infection. To determine whether the presence of polyfunctional CD8+ T cell responses distinguishes protected and unprotected monkeys in a live attenuated lentivirus model, SIV Gag peptide-specific CD8+ T cell responses of simian HIV (SHIV) 89.6-vaccinated, SIVmac239-challenged rhesus macaques were compared in two monkeys that controlled challenge virus replication and two that did not. The ratio of Bcl-2+ Gag-specific CD8+ T cells to caspase-3+ Gag-specific CD8+ T cells was higher in the vaccinated-protected animals compared with unprotected monkeys. In addition, polyfunctional SIV-specific CD8+ T cells were consistently detected through 12 wk postchallenge in the protected animals but not in the unprotected animals. In the unprotected monkeys, there was an increased frequency of CD8+ T cells expressing markers associated with effector memory T cells. Further, there was increased annexin V expression in central memory T cells of the unprotected animals before challenge. Thus, monkeys that control viral replication after live attenuated SHIV infection have polyfunctional SIV-specific CD8+ T cells with an increased survival potential. Importantly, the differences in the nature of the SIV-specific CD8+ T cell response in the protected and unprotected animals are present during acute stages postchallenge, before different antigenic levels are established. Thus, the polyfunctional capacity and increased survival potential of CD8+ SIV-specific T cells may account for live attenuated, SHIV89.6-mediated protection from uncontrolled SIV replication.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Produtos do Gene gag/imunologia , Infecções por Lentivirus/imunologia , Macaca mulatta/imunologia , Vírus da Imunodeficiência Símia/imunologia , Vírus da Imunodeficiência Símia/patogenicidade , Replicação Viral , Animais , Apoptose , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Suscetibilidade a Doenças , Memória Imunológica/imunologia , Infecções por Lentivirus/sangue , Infecções por Lentivirus/patologia , Infecções por Lentivirus/virologia , Macaca mulatta/virologia , Masculino , Fenótipo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Vírus de RNA/metabolismo , Vírus da Imunodeficiência Símia/química
17.
Am J Physiol Lung Cell Mol Physiol ; 292(5): L1233-40, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17220371

RESUMO

Pneumonia is a major complication of human immunodeficiency virus (HIV) pathogenesis but it develops only after prolonged infection. We used the macaque model to explore a hypothesis that the disease is a two-stage process, the first stage being establishment of the viral infection in the lung and the second being amplification of virus replication by host factors induced by chemical agents or opportunistic pathogens in the lung. Bleomycin, a chemical known to induce diffuse alveolar damage and pulmonary fibrosis with accumulation of macrophages and a rich T helper type 2 (Th2) cytokine environment, was inoculated intratracheally into five of eight SHIV 89.6P-infected macaques and into one uninfected macaque. Three additional simian HIV (SHIV)-infected macaques without bleomycin treatment served as untreated virus controls. Although none of the animals became clinically ill, bleomycin induced classical host responses in the lungs of all the treated, virus-infected macaques. There was enhanced production of the chemokine, monocyte chemotactic protein-1 (MCP-1), that had previously been shown to cause enhanced replication of the virus. Four of the five treated animals developed more productive SHIV infection in the lungs compared with the infected untreated animals. Enhanced virus replication was found primarily in infiltrating macrophages. Enhanced replication of the virus in the lungs was associated with host factors induced by the drug and supported the hypothesis for a two-stage process of pulmonary pathogenesis.


Assuntos
Bleomicina/farmacologia , Infecções por Lentivirus/patologia , Pulmão/virologia , Vírus da Imunodeficiência Símia/fisiologia , Replicação Viral/efeitos dos fármacos , Animais , Imuno-Histoquímica , Macaca , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Macrófagos/virologia , Vírus da Imunodeficiência Símia/efeitos dos fármacos
18.
Comp Immunol Microbiol Infect Dis ; 26(1): 1-15, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12602682

RESUMO

Bovine immunodeficiency virus (BIV) in Holstein cows was associated with morphologic evidence of lymphoid organ deficiency. Cows were subjected to normal management practices including parturition and lactation without adverse environmental stresses. During the clinical disease process there was marked weight loss and wasting with frequent and severe concurrent infections. Lymphoid follicular hyperplasia and dysplasia in lymph nodes, and hypertrophy and hyperplasia in hemal lymph nodes were characteristics of the lymphoid tissues. Atrophy of lymphoid cell compartments with depletion of lymphocytes and a lymphocytic lymphoid folliculitis were components of the lymphoid system pathology. The nodal tissue lesions resembled those observed in feline, simian, and human lentiviral disease. A functional correlation with immune system deficiency was the development of multiple bacterial infections which failed to resolve after appropriate therapy. The BIV-associated disease syndrome in dairy cows may be useful as a model system for investigation of the pathogenesis of the lymphoid organ changes that occur in humans and animals with lentiviral infection.


Assuntos
Doenças dos Bovinos/patologia , Vírus da Imunodeficiência Bovina/patogenicidade , Infecções por Lentivirus/veterinária , Tecido Linfoide/patologia , Animais , Bovinos , Doenças dos Bovinos/imunologia , Doenças dos Bovinos/virologia , Modelos Animais de Doenças , Feminino , Infecções por Lentivirus/imunologia , Infecções por Lentivirus/patologia , Linfonodos/citologia , Linfonodos/patologia , Linfócitos/imunologia , Neutrófilos/imunologia , Infecções Oportunistas/veterinária
19.
Am J Pathol ; 161(3): 813-22, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12213709

RESUMO

Neurological disease associated with lentiviral infection occurs mainly as a consequence of primary replication of the virus or a combination of the virus infection and replication of opportunistic pathogens in the central nervous system. Recent studies have shown that whereas the disease can be caused by CCR5 tropic viruses alone, its induction by CXCR4 (X4) tropic viruses occurred usually in association with infections caused by opportunistic pathogens and in the presence of a Th2 cytokine, interleukin (IL)-4.(1,2) Further, X4-mediated neurological disease developed preferentially in rhesus compared to pig-tailed macaques. Because macrophages are the target cells for lentiviral infection in the brain and because macrophage chemoattractant protein (MCP)-1 is one of the major chemokines that is closely associated with acquired immune deficiency syndrome (AIDS) dementia, we tested for correlations between MCP-1 production and virus tropism in macrophages from the two species of macaques. The studies showed that the higher susceptibility of rhesus macaques to X4 virus-mediated encephalitis correlated with heightened production of virus and MCP-1 in cultured macrophages from this species and that these effects were further enhanced with treatment with IL-4. However, the latter effect was restricted to macrophages infected with X4 viruses. IL-4 may therefore be a basic requirement for X4 viruses to cause central nervous system disease.


Assuntos
Adjuvantes Imunológicos/farmacologia , Quimiocina CCL2/biossíntese , Interleucina-4/farmacologia , Infecções por Lentivirus/metabolismo , Macrófagos/metabolismo , Macrófagos/virologia , Receptores CCR5/metabolismo , Receptores CXCR4/metabolismo , Animais , Células Cultivadas , Lentivirus/fisiologia , Infecções por Lentivirus/patologia , Macaca , Replicação Viral/efeitos dos fármacos
20.
J Virol ; 76(6): 2622-33, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11861828

RESUMO

The release of neurotoxins by activated brain macrophages or microglia is one mechanism proposed to contribute to the development of neurological disease following infection by lentiviruses, including feline immunodeficiency virus (FIV). Since molecular diversity in the lentiviral envelope gene influences the expression of host molecules implicated in neuronal injury, the role of the envelope sequence in FIV neuropathogenesis was investigated by using the neurovirulent FIV strain V1CSF, the nonneurovirulent strain Petaluma, and a chimera (FIVCh) containing the V1CSF envelope gene in a Petaluma background. All three viruses replicated in primary feline macrophages with equal efficiency, but conditioned medium from V1CSF- or FIVCh-infected cells was significantly more neurotoxic than medium from Petaluma-infected cultures (P < 0.001) and could be attenuated in a dose-dependent manner by treatment with either the matrix metalloproteinase (MMP) inhibitor prinomastat (PMT) or function-blocking antibodies to MMP-2. Although FIV sequences were detectable by PCR in brain tissue from neonatal cats infected with each of the viral strains, immunohistochemistry revealed increased astrogliosis and macrophage activation in the brains of V1CSF- and FIVCh-infected cats relative to the other groups, together with elevated markers of neuronal stress that included morphological changes and increased c-fos immunoreactivity. Similarly, MMP-2, but not MMP-9, mRNA and protein expression was increased in brain tissues of V1CSF- and FIVCh-infected cats relative to Petaluma-infected animals (P < 0.01). Infection with V1CSF or FIVCh was also associated with greater CD4(+) cell depletion (P < 0.001) and neurodevelopmental delays (P < 0.005), than in Petaluma-infected animals; these deficits improved following PMT therapy. These findings indicated that diversity in the envelope gene sequence influenced the neurovirulence exhibited by FIV both in vitro and in vivo, possibly through a mechanism involving the differential induction of MMP-2.


Assuntos
Viroses do Sistema Nervoso Central/virologia , Regulação Viral da Expressão Gênica , Genes env/genética , Vírus da Imunodeficiência Felina/patogenicidade , Infecções por Lentivirus/virologia , Animais , Animais Recém-Nascidos , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/virologia , Gatos , Viroses do Sistema Nervoso Central/patologia , Viroses do Sistema Nervoso Central/fisiopatologia , Feminino , Humanos , Vírus da Imunodeficiência Felina/genética , Recém-Nascido , Infecções por Lentivirus/patologia , Infecções por Lentivirus/fisiopatologia , Macrófagos/virologia , Metaloproteinases da Matriz/metabolismo , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Gravidez , Células Tumorais Cultivadas , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA