Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 203
Filtrar
1.
mBio ; 11(5)2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32934084

RESUMO

Bats are primary reservoirs for multiple lethal human viruses, such as Ebola, Nipah, Hendra, rabies, severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome-related coronavirus (MERS-CoV), and, most recently, SARS-CoV-2. The innate immune systems of these immensely abundant, anciently diverged mammals remain insufficiently characterized. While bat genomes contain many endogenous retroviral elements indicative of past exogenous infections, little is known about restrictions to extant retroviruses. Here, we describe a major postentry restriction in cells of the yinpterochiropteran bat Pteropus alecto Primate lentiviruses (HIV-1, SIVmac) were potently blocked at early life cycle steps, with up to 1,000-fold decreases in infectivity. The block was specific, because nonprimate lentiviruses such as equine infectious anemia virus and feline immunodeficiency virus were unimpaired, as were foamy retroviruses. Interspecies heterokaryons demonstrated a dominant block consistent with restriction of incoming viruses. Several features suggested potential TRIM5 (tripartite motif 5) or myxovirus resistance protein 2 (MX2) protein restriction, including postentry action, cyclosporine sensitivity, and reversal by capsid cyclophilin A (CypA) binding loop mutations. Viral nuclear import was significantly reduced, and this deficit was substantially rescued by cyclosporine treatment. However, saturation with HIV-1 virus-like particles did not relieve the restriction at all. P. alecto TRIM5 was inactive against HIV-1 although it blocked the gammaretrovirus N-tropic murine leukemia virus. Despite major divergence in a critical N-terminal motif required for human MX2 activity, P. alecto MX2 had anti-HIV activity. However, this did not quantitatively account for the restriction and was independent of and synergistic with an additional CypA-dependent restriction. These results reveal a novel, specific restriction to primate lentiviruses in the Pteropodidae and advance understanding of bat innate immunity.IMPORTANCE The COVID-19 pandemic suggests that bat innate immune systems are insufficiently characterized relative to the medical importance of these animals. Retroviruses, e.g., HIV-1, can be severe pathogens when they cross species barriers, and bat restrictions corresponding to retroviruses are comparatively unstudied. Here, we compared the abilities of retroviruses from three genera (Lentivirus, Gammaretrovirus, and Spumavirus) to infect cells of the large fruit-eating bat P. alecto and other mammals. We identified a major, specific postentry restriction to primate lentiviruses. HIV-1 and SIVmac are potently blocked at early life cycle steps, but nonprimate lentiviruses and foamy retroviruses are entirely unrestricted. Despite acting postentry and in a CypA-dependent manner with features reminiscent of antiretroviral factors from other mammals, this restriction was not saturable with virus-like particles and was independent of P. alecto TRIM5, TRIM21, TRIM22, TRIM34, and MX2. These results identify a novel restriction and highlight cyclophilin-capsid interactions as ancient species-specific determinants of retroviral infection.


Assuntos
Quirópteros/imunologia , Gammaretrovirus/imunologia , Imunidade Inata/imunologia , Lentivirus de Primatas/imunologia , Spumavirus/imunologia , Células 3T3 , Animais , Aotidae , Gatos , Linhagem Celular , Quirópteros/virologia , Ciclofilina A/metabolismo , Furões , Gammaretrovirus/crescimento & desenvolvimento , Células HEK293 , Humanos , Lentivirus de Primatas/crescimento & desenvolvimento , Camundongos , Interferência de RNA , RNA Interferente Pequeno/genética , Spumavirus/crescimento & desenvolvimento , Proteínas com Motivo Tripartido/metabolismo
2.
Mol Ther ; 25(5): 1132-1141, 2017 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-28366768

RESUMO

Prior to the first successful bone marrow transplant in 1968, patients born with severe combined immunodeficiency (SCID) invariably died. Today, with a widening availability of newborn screening, major improvements in the application of allogeneic procedures, and the emergence of successful hematopoietic stem and progenitor cell (HSC/P) gene therapy, the majority of these children can be identified and cured. Here, we trace key steps in the development of clinical gene therapy for SCID and other primary immunodeficiencies (PIDs), and review the prospects for adoption of new targets and technologies.


Assuntos
Adenosina Desaminase/genética , Terapia Genética/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Síndromes de Imunodeficiência/terapia , Imunodeficiência Combinada Severa/terapia , Adenosina Desaminase/deficiência , Adenosina Desaminase/imunologia , Ensaios Clínicos como Assunto , Gammaretrovirus/genética , Gammaretrovirus/imunologia , Expressão Gênica , Terapia Genética/história , Vetores Genéticos/química , Vetores Genéticos/imunologia , História do Século XX , História do Século XXI , Humanos , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Síndromes de Imunodeficiência/patologia , Lentivirus/genética , Lentivirus/imunologia , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/imunologia , Imunodeficiência Combinada Severa/patologia , Transplante Homólogo
3.
Mol Ther ; 25(5): 1142-1154, 2017 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-28377044

RESUMO

ß-Thalassemia and sickle cell disease (SCD) are the world's two most widely disseminated hereditary hemoglobinopathies. ß-Thalassemia originated in the Mediterranean, Middle Eastern, and Asian regions, and SCD originated in central Africa. However, subsequent population migration means that these two diseases are now global and thus constitute a growing health problem in many countries. Despite remarkable improvements in medical care for patients with ß-hemoglobinopathies, there is still only one definitive treatment option: allogeneic hematopoietic stem cell (HSC) transplantation. The development of gene therapy for ß-hemoglobinopathies has been justified by (1) the limited availability of human leukocyte antigen (HLA)-identical donors, (2) the narrow window of application of HSC transplantation to the youngest patients, and (3) recent advances in HSC-based gene therapy. The huge ongoing efforts in translational medicine and the high number of related publications show that gene therapy has the potential to become the treatment of choice for patients who lack either an HLA genoidentical sibling or an alternative, medically acceptable donor. In this dynamic scientific context, we first summarize the main steps toward clinical translation of this therapeutic approach and then discuss novel lentiviral- and genome editing-based treatment strategies for ß-hemoglobinopathies.


Assuntos
Anemia Falciforme/terapia , Edição de Genes/métodos , Terapia Genética/métodos , Transplante de Células-Tronco Hematopoéticas , Globinas beta/genética , Talassemia beta/terapia , Anemia Falciforme/genética , Anemia Falciforme/metabolismo , Anemia Falciforme/patologia , Gammaretrovirus/genética , Gammaretrovirus/imunologia , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/imunologia , Antígenos HLA , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Lentivirus/genética , Lentivirus/imunologia , Mutação , Doadores de Tecidos , Transplante Homólogo , Globinas beta/metabolismo , Talassemia beta/genética , Talassemia beta/metabolismo , Talassemia beta/patologia
4.
Hum Gene Ther ; 27(2): 108-16, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26790362

RESUMO

More than 20 years ago, X-linked severe combined immunodeficiency (SCID-X1) appeared to be the best condition to test the feasibility of hematopoietic stem cell gene therapy. The seminal SCID-X1 clinical studies, based on first-generation gammaretroviral vectors, demonstrated good long-term immune reconstitution in most treated patients despite the occurrence of vector-related leukemia in a few of them. This gene therapy has successfully enabled correction of the T cell defect. Natural killer and B cell defects were only partially restored, most likely due to the absence of a conditioning regimen. The success of these pioneering trials paved the way for the extension of gene-based treatment to many other diseases of the hematopoietic system, but the unfortunate serious adverse events led to extensive investigations to define the retrovirus integration profiles. This review puts into perspective the clinical experience of gene therapy for SCID-X1, with the development and implementation of new generations of safer vectors such as self-inactivating gammaretroviral or lentiviral vectors as well as major advances in integrome knowledge.


Assuntos
Gammaretrovirus/genética , Terapia Genética/métodos , Vetores Genéticos/química , Lentivirus/genética , Retroviridae/genética , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/terapia , Linfócitos B/imunologia , Linfócitos B/patologia , Ensaios Clínicos como Assunto , Gammaretrovirus/imunologia , Vetores Genéticos/imunologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Lentivirus/imunologia , Segurança do Paciente , Retroviridae/imunologia , Linfócitos T/imunologia , Linfócitos T/patologia , Integração Viral/genética , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/genética , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/imunologia , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/patologia
5.
J Virol ; 88(4): 2349-53, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24335302

RESUMO

Interferon regulatory factor (IRF) regulation of the type I interferon response has not been extensively explored in murine retroviral infections. IRF-3(-/-) and select IRF-3/7(-/-) mice were resistant to LP-BM5-induced pathogenesis. However, further analyses strongly suggested that resistance could be attributed to strain 129-specific contamination of the known retrovirus resistance gene Fv1. Therefore, caution should be taken when interpreting phenotypes observed in these knockout mice, as strain 129-derived genetic polymorphisms may explain observed differences.


Assuntos
Modelos Animais de Doenças , Gammaretrovirus/imunologia , Fator Regulador 3 de Interferon/genética , Fator Regulador 7 de Interferon/genética , Interferon Tipo I/imunologia , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Síndrome de Imunodeficiência Adquirida Murina/fisiopatologia , Animais , Eletroforese , Gammaretrovirus/genética , Camundongos , Camundongos Knockout , Síndrome de Imunodeficiência Adquirida Murina/virologia , Proteínas/genética , Proteínas/imunologia , Especificidade da Espécie , Estatísticas não Paramétricas
6.
Curr Opin Virol ; 3(6): 657-63, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23992668

RESUMO

Laboratory mice carry three host range groups of gammaretroviruses all of which are linked to leukemia induction. Although polytropic mouse leukemia viruses (P-MLVs) are generally recognized as the proximate cause of MLV-induced leukemias in laboratory mice, wild mice that carry only endogenous P-MLVs do not produce infectious virus and are not prone to disease; these mice carry the permissive XPR1 retroviral receptor and an attenuated variant of the retroviral restriction factor, APOBEC3. In contrast, Eurasian mice carrying ecotropic and xenotropic MLVs have evolved multiple restrictive XPR1 variants, other factors that interfere with MLV entry, and more effectively antiviral variants of APOBEC3. These different antiviral restrictions in Mus musculus subspecies suggest that the different virus types found in these natural populations may pose different but largely uncharacterized survival risks in their host subspecies.


Assuntos
Citidina Desaminase/metabolismo , Gammaretrovirus/imunologia , Interações Hospedeiro-Patógeno , Receptores Acoplados a Proteínas G/metabolismo , Receptores Virais/metabolismo , Animais , Citidina Desaminase/imunologia , Gammaretrovirus/fisiologia , Regulação Viral da Expressão Gênica , Camundongos , Internalização do Vírus , Replicação Viral , Receptor do Retrovírus Politrópico e Xenotrópico
7.
Hum Vaccin Immunother ; 9(3): 462-70, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23249763

RESUMO

The induction of neutralizing antibodies is a promising way to prevent retrovirus infections. Neutralizing antibodies are mainly directed against the envelope proteins, which consist of two molecules, the surface envelope (SU) protein and the transmembrane envelope (TM) protein. Antibodies broadly neutralizing the human immunodeficiencvy virus-1 (HIV-1) and binding to the TM protein gp41 of the virus have been isolated from infected individuals. Their epitopes are located in the membrane proximal external region (MPER). Since there are difficulties to induce such neutralizing antibodies as basis for an effective AIDS vaccine, we performed a comparative analysis immunising with the TM proteins of different viruses from the family Retroviridae. Both subfamilies, the Orthoretrovirinae and the Spumaretrovirinae were included. In this study, the TM proteins of three gammaretroviruses including (1) the porcine endogenous retrovirus (PERV), (2) the Koala retrovirus (KoRV), (3) the feline leukemia virus (FeLV), of two lentiviruses, HIV-1, HIV-2, and of two spumaviruses, the feline foamy virus (FFV) and the primate foamy virus (PFV) were used for immunisation. Whereas in all immunisation studies binding antibodies were induced, neutralizing antibodies were only found in the case of the gammaretroviruses. The induced antibodies were directed against the MPER and the fusion peptide proximal region (FPPR) of their TM proteins; however only the antibodies against the MPER were neutralizing. Most importantly, the epitopes in the MPER were localized in the same position as the epitopes of the antibodies broadly neutralizing HIV-1 in the TM protein gp41 of HIV-1, indicating that the MPER is an effective target for the neutralization of retroviruses.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Gammaretrovirus/imunologia , Lentivirus/imunologia , Spumavirus/imunologia , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Mapeamento de Epitopos , Proteínas do Envelope Viral/administração & dosagem , Vacinas Virais/administração & dosagem
8.
J Immunother ; 35(5): 430-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22576348

RESUMO

The clinical application of interleukin-12 (IL-12) has been hindered by the toxicity associated with its systemic administration. To potentially overcome this problem, we developed a promoter designed to direct IL-12 expression within the tumor environment using an inducible composite promoter containing binding motifs for the nuclear factor of activated T cells (NFAT) linked to a minimal IL-2 promoter. In this study, the NFAT promoter was coupled to a single-chain human IL-12 gene and inserted into 2 γ-retroviral self-inactivating vectors (SERS.NFAT.hIL12 and SERS.NFAT.hIL12.PA2) and 1 γ-retroviral vector (MSGV1.NFAT.hIL.12 PA2). Peripheral blood lymphocytes (PBLs) were double transduced with an antigen-specific T-cell receptor and the 3 NFAT.hIL12 vectors. Evaluation of inducible IL-12 expression, transduction efficiency, and vector production considerations led to the choice of the MSGV1.NFAT.hIL12.PA2 vector for clinical application. MSGV1.NFAT.hIL12.PA2 PG13 retroviral vector producer cell clones were screened by transduction of tumor antigen-specific PBLs. On the basis of expression studies in PBL, clone D3 was chosen to produce clinical-grade viral vector supernatant and was demonstrated to efficiently transduce young tumor-infiltrating lymphocytes (TIL). The vector-transduced young TIL with known tumor recognition demonstrated specific inducible IL-12 production after coculture with HLA-matched tumor targets and had augmented effector function as demonstrated by increased IFN-γ secretion. These results support the clinical application of adoptive transfer of young TIL engineered with the NFAT.hIL12 vector as a new approach for cancer immunotherapy.


Assuntos
Gammaretrovirus/imunologia , Interleucina-12/biossíntese , Neoplasias/terapia , Transferência Adotiva , Técnicas de Cocultura , Gammaretrovirus/genética , Expressão Gênica , Vetores Genéticos , Antígenos HLA/imunologia , Humanos , Imunoterapia , Interferon gama/biossíntese , Interferon gama/imunologia , Interleucina-12/genética , Interleucina-12/imunologia , Ativação Linfocitária , Linfócitos do Interstício Tumoral/imunologia , Neoplasias/imunologia , Regiões Promotoras Genéticas , Receptores de Antígenos de Linfócitos T/biossíntese , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Transdução Genética
10.
Hum Vaccin ; 7 Suppl: 4-9, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21266839

RESUMO

Although the development of an effective vaccine is the main goal in the fight against AIDS, all attempts by numerous laboratories to develop a vaccine have failed so far. In addition, it is still unclear whether cytotoxic T cells or neutralizing antibodies or both should be induced. The major advantage of neutralizing antibodies is their ability to prevent infection and subsequent integration of the provirus into the cellular genome where it may persist in a form invisible to the immune system. Broadly neutralizing antibodies have been found in HIV infected individuals, including antibodies directed against a highly conserved region in the membrane proximal external region (MPER) of the transmembrane envelope (TM) protein gp41 of HIV-1. We successfully induced neutralizing antibodies against different gammaretroviruses by immunization with their respective TM proteins. These antibodies recognized epitopes not only in the MPER but also in the fusion peptide proximal region of the TM protein. In the case of feline leukaemia virus (FeLV), these antibodies protected cats from antigenemia following challenge. To understand the mechanism of neutralization, the interactions between neutralizing antibodies and their corresponding epitopes in the TM protein of gammaretroviruses and HIV-1 were analysed. These data may help to design antigens able to induce specific broadly neutralizing antibodies.


Assuntos
Vacinas contra a AIDS/imunologia , Síndrome da Imunodeficiência Adquirida/prevenção & controle , Síndrome da Imunodeficiência Adquirida/terapia , Anticorpos Neutralizantes/imunologia , Anticorpos Anti-HIV/imunologia , Proteína gp41 do Envelope de HIV/imunologia , Animais , Gammaretrovirus/imunologia , Humanos
11.
J Virol ; 85(3): 1205-13, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21084477

RESUMO

A novel gammaretrovirus, xenotropic murine leukemia virus-related virus (XMRV), has been identified in patients with prostate cancer and in patients with chronic fatigue syndromes. Standard Mus musculus laboratory mice lack a functional XPR1 receptor for XMRV and are therefore not a suitable model for the virus. In contrast, Gairdner's shrew-mice (Mus pahari) do express functional XPR1. To determine whether Mus pahari could serve as a model for XMRV, primary Mus pahari fibroblasts and mice were infected with cell-free XMRV. Infection of cells in vitro resulted in XMRV Gag expression and the production of XMRV virions. After intraperitoneal injection of XMRV into Mus pahari mice, XMRV proviral DNA could be detected in spleen, blood, and brain. Intravenous administration of a green fluorescent protein (GFP) vector pseudotyped with XMRV produced GFP(+) CD4(+) T cells and CD19(+) B cells. Mice mounted adaptive immune responses against XMRV, as evidenced by the production of neutralizing and Env- and Gag-specific antibodies. Prominent G-to-A hypermutations were also found in viral genomes isolated from the spleen, suggesting intracellular restriction of XMRV infection by APOBEC3 in vivo. These data demonstrate infection of Mus pahari by XMRV, potential cell tropism of the virus, and immunological and intracellular restriction of virus infection in vivo. These data support the use of Mus pahari as a model for XMRV pathogenesis and as a platform for vaccine and drug development against this potential human pathogen.


Assuntos
Modelos Animais de Doenças , Gammaretrovirus/patogenicidade , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Virais/genética , Receptores Virais/metabolismo , Infecções por Retroviridae/virologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Antígenos CD19/análise , Linfócitos B/química , Linfócitos B/virologia , Sangue/virologia , Encéfalo/virologia , Linfócitos T CD4-Positivos/virologia , Células Cultivadas , Fibroblastos/virologia , Gammaretrovirus/imunologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/patologia , Baço/virologia , Coloração e Rotulagem/métodos , Tropismo Viral , Receptor do Retrovírus Politrópico e Xenotrópico
12.
Retrovirology ; 7: 68, 2010 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-20716359

RESUMO

BACKGROUND: Xenotropic Murine Leukemia Virus-related Virus (XMRV) is a human gammaretrovirus recently identified in prostate cancer tissue and in lymphocytes of patients with chronic fatigue syndrome. To establish the etiologic role of XMRV infection in human disease requires large scale epidemiologic studies. Development of assays to detect XMRV-specific antibodies would greatly facilitate such studies. However, the nature and kinetics of the antibody response to XMRV infection have yet to be determined. RESULTS: Three rhesus macaques were infected with XMRV to determine the dynamics of the antibody responses elicited by infection with XMRV. All macaques developed antibodies to XMRV during the second week of infection, and the predominant responses were to the envelope protein gp70, transmembrane protein p15E, and capsid protein p30. In general, antibody responses to gp70 and p15E appeared early with higher titers than to p30, especially in the early period of seroconversion. Antibodies to gp70, p15E and p30 persisted to 158 days and were substantially boosted by re-infection, thus, were identified as useful serologic markers. Three high-throughput prototype assays were developed using recombinant proteins to detect antibodies to these viral proteins. Both gp70 and p15E prototype assays demonstrated 100% sensitivity by detecting all Western blot (WB) positive serial bleeds from the XMRV-infected macaques and good specificity (99.5-99.9%) with blood donors. Seroconversion sensitivity and specificity of the p30 prototype assay were 92% and 99.4% respectively. CONCLUSIONS: This study provides the first demonstration of seroconversion patterns elicited by XMRV infection. The nature and kinetics of antibody responses to XMRV in primates were fully characterized. Moreover, key serologic markers useful for detection of XMRV infection were identified. Three prototype immunoassays were developed to detect XMRV-specific antibodies. These assays demonstrated good sensitivity and specificity; thus, they will facilitate large scale epidemiologic studies of XMRV infection in humans.


Assuntos
Anticorpos Antivirais/sangue , Gammaretrovirus/isolamento & purificação , Infecções por Retroviridae/diagnóstico , Infecções por Retroviridae/epidemiologia , Virologia/métodos , Animais , Modelos Animais de Doenças , Estudos Epidemiológicos , Feminino , Gammaretrovirus/imunologia , Humanos , Imunoensaio/métodos , Macaca mulatta , Masculino , Proteínas Recombinantes/imunologia , Infecções por Retroviridae/imunologia , Sensibilidade e Especificidade , Proteínas Estruturais Virais/imunologia
13.
Nat Rev Urol ; 7(7): 392-402, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20517289

RESUMO

Xenotropic murine leukemia virus-related virus (XMRV) is an authentic, newly recognized human retrovirus first identified in prostate cancer tissues from men with a deficiency in the innate immunity gene RNASEL. At present, studies have detected XMRV at widely different rates in prostate cancer cases (0-27%) and in patients with chronic fatigue syndrome (CFS; 0-67%). Indirect or direct modes of carcinogenesis by XMRV have been suggested depending on whether the virus was found in stroma or malignant epithelium. Viral replication in the prostate might be affected by androgens, which stimulate XMRV through a transcriptional enhancer site in viral DNA. By contrast, host restriction factors, such as APOBEC3 and tetherin, inhibit virus replication. Immune dysfunction mediated by XMRV has been suggested as a possible factor in CFS. Recent studies show that some existing antiretroviral drugs suppress XMRV infections and diagnostic assays are under development. Although other retroviruses of the same genus as XMRV (gammaretroviruses) cause cancer and neurological disease in animals, whether XMRV is a cause of either prostate cancer or CFS remains unknown. Emerging science surrounding XMRV is contributing to our knowledge of retroviral infections while focusing intense interest on two major human diseases.


Assuntos
Síndrome de Fadiga Crônica/virologia , Neoplasias da Próstata/virologia , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/virologia , Antirretrovirais/farmacologia , Antirretrovirais/uso terapêutico , Síndrome de Fadiga Crônica/imunologia , Síndrome de Fadiga Crônica/terapia , Feminino , Gammaretrovirus/efeitos dos fármacos , Gammaretrovirus/imunologia , Gammaretrovirus/patogenicidade , Humanos , Vírus da Leucemia Murina/efeitos dos fármacos , Vírus da Leucemia Murina/imunologia , Vírus da Leucemia Murina/patogenicidade , Masculino , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/terapia , Retroviridae/efeitos dos fármacos , Retroviridae/imunologia , Retroviridae/patogenicidade , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/terapia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/terapia
14.
J Virol ; 84(11): 5719-29, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20335265

RESUMO

Xenotropic murine leukemia virus-related virus (XMRV), a gammaretrovirus, has been isolated from human prostate cancer tissue and from activated CD4(+) T cells and B cells of patients with chronic fatigue syndrome, suggesting an association between XMRV infection and these two diseases. Since APOBEC3G (A3G) and APOBEC3F (A3F), which are potent inhibitors of murine leukemia virus and Vif-deficient human immunodeficiency virus type 1 (HIV-1), are expressed in human CD4(+) T cells and B cells, we sought to determine how XMRV evades suppression of replication by APOBEC3 proteins. We found that expression of A3G, A3F, or murine A3 in virus-producing cells resulted in their virion incorporation, inhibition of XMRV replication, and G-to-A hypermutation of the viral DNA with all three APOBEC3 proteins. Quantitation of A3G and A3F mRNAs indicated that, compared to the human T-cell lines CEM and H9, prostate cell lines LNCaP and DU145 exhibited 50% lower A3F mRNA levels, whereas A3G expression in 22Rv1, LNCaP, and DU145 cells was nearly undetectable. XMRV proviral genomes in LNCaP and DU145 cells were hypermutated at low frequency with mutation patterns consistent with A3F activity. XMRV proviral genomes were extensively hypermutated upon replication in A3G/A3F-positive T cells (CEM and H9), but not in A3G/A3F-negative cells (CEM-SS). We also observed that XMRV replication was susceptible to the nucleoside reverse transcriptase (RT) inhibitors zidovudine (AZT) and tenofovir and the integrase inhibitor raltegravir. In summary, the establishment of XMRV infection in patients may be dependent on infection of A3G/A3F-deficient cells, and cells expressing low levels of A3G/A3F, such as prostate cancer cells, may be ideal producers of infectious XMRV. Furthermore, the anti-HIV-1 drugs AZT, tenofovir, and raltegravir may be useful for treatment of XMRV infection.


Assuntos
Antivirais/farmacologia , Citidina Desaminase/imunologia , Gammaretrovirus/imunologia , Vírus da Leucemia Murina/imunologia , Neoplasias da Próstata/virologia , Infecções por Retroviridae/imunologia , Infecções Tumorais por Vírus/imunologia , Adenina/análogos & derivados , Adenina/farmacologia , Animais , Linhagem Celular Tumoral , Citidina Desaminase/genética , Humanos , Masculino , Camundongos , Mutação , Organofosfonatos/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Pirrolidinonas/farmacologia , Raltegravir Potássico , Infecções por Retroviridae/tratamento farmacológico , Tenofovir , Infecções Tumorais por Vírus/tratamento farmacológico , Zidovudina/farmacologia
15.
Science ; 326(5952): 585-9, 2009 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-19815723

RESUMO

Chronic fatigue syndrome (CFS) is a debilitating disease of unknown etiology that is estimated to affect 17 million people worldwide. Studying peripheral blood mononuclear cells (PBMCs) from CFS patients, we identified DNA from a human gammaretrovirus, xenotropic murine leukemia virus-related virus (XMRV), in 68 of 101 patients (67%) as compared to 8 of 218 (3.7%) healthy controls. Cell culture experiments revealed that patient-derived XMRV is infectious and that both cell-associated and cell-free transmission of the virus are possible. Secondary viral infections were established in uninfected primary lymphocytes and indicator cell lines after their exposure to activated PBMCs, B cells, T cells, or plasma derived from CFS patients. These findings raise the possibility that XMRV may be a contributing factor in the pathogenesis of CFS.


Assuntos
Síndrome de Fadiga Crônica/virologia , Gammaretrovirus/isolamento & purificação , Leucócitos Mononucleares/virologia , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/virologia , Animais , Anticorpos Antivirais/sangue , Linfócitos B/imunologia , Linfócitos B/virologia , Sequência de Bases , Linhagem Celular , Linhagem Celular Tumoral , Técnicas de Cocultura , DNA/genética , Gammaretrovirus/genética , Gammaretrovirus/imunologia , Gammaretrovirus/fisiologia , Produtos do Gene env/análise , Produtos do Gene gag/análise , Genoma Viral , Humanos , Ativação Linfocitária , Masculino , Camundongos , Dados de Sequência Molecular , Neoplasias da Próstata/virologia , Infecções por Retroviridae/epidemiologia , Infecções por Retroviridae/transmissão , Linfócitos T/imunologia , Linfócitos T/virologia , Infecções Tumorais por Vírus/epidemiologia , Infecções Tumorais por Vírus/transmissão
17.
J Virol ; 82(13): 6120-9, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18417580

RESUMO

Cells of Mus minutoides, an African pygmy mouse of the subgenus Nannomys, are susceptible to ecotropic Moloney and Friend mouse leukemia viruses (MLVs) but not to AKV-type MLVs. Transfected MA139 ferret cells expressing the mCAT-1 cell surface receptor, with the minCAT-1 substitutions K222Q and V233L, did not restrict AKV MLV. The resistance of M. minutoides cells to AKV MLV was not relieved by inhibitors of glycosylation or by the introduction of NIH 3T3 mCAT-1. Resistance is thus not mediated by receptor sequence variation, expression level, or glycosylation. M. minutoides cells are also infectible with LacZ pseudotypes having AKV Env and Moloney MLV (MoMLV) Gag proteins, further indicating that AKV Env sequence variations do not contribute to the observed block. The pattern of virus resistance in M. minutoides differs from that of the known variants of the Fv1 postentry resistance gene; M. minutoides is equally resistant to N-, B-, and NR-tropic AKV viruses and is equally susceptible to NR- and NB-tropic Friend MLVs. This novel resistance blocks replication before reverse transcription, whereas Fv1 generally restricts replication after reverse transcription; M. minutoides cells produce 2-long-terminal-repeat viral DNA circles and linear viral DNA after infection with MoMLV but not with AKV MLV. Analysis of MoMLV-AKV MLV chimeras determined that the target of resistance is in the virus capsid gene. Mutagenesis demonstrated that restriction is mediated by two amino acid substitutions, H117L and A110R; substitutions at these sites can also be targeted by the resistance genes Fv1 and TRIM5alpha. M. minutoides cells thus have a novel postentry resistance to AKV MLVs.


Assuntos
Gammaretrovirus/imunologia , Imunidade Inata/imunologia , Infecções por Retroviridae/imunologia , Infecções Tumorais por Vírus/imunologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Transportador 1 de Aminoácidos Catiônicos/genética , Linhagem Celular , Produtos do Gene gag/genética , Imunidade Inata/genética , Camundongos , Dados de Sequência Molecular , Filogenia , Análise de Sequência de DNA
18.
Biotechnol Bioeng ; 101(2): 357-68, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18435481

RESUMO

Development of methods to engineer gamma-retroviral vectors capable of transducing target cells in a cell-specific manner could impact the future of the clinical application of gene therapy as well as the understanding of the biology of transfer gene vectors. Two molecular events are critical for controlling the entry of gamma-retroviral vectors to target cells: binding to cell-surface receptors and the subsequent fusion of viral vector membrane and cellular membrane. In this report, we evaluated a method to incorporate a membrane-bound antibody and a fusogenic molecule to provide binding and fusion functions respectively, into gamma-retroviral vectors for targeted gene delivery. An anti-CD20 antibody and a fusogenic protein derived from Sindbis virus glycoprotein could be efficiently co-displayed on the surface of viral vectors. Vectors bearing anti-CD20 antibody conferred their binding specificity to cells expressing CD20. Enhanced in vitro transduction towards CD20-expressing cells was observed for gamma-retroviral vectors displaying both an antibody and a fusogen. We found that the biological activity of the fusogen played an important role on the efficiency of such a targeting strategy and were able to engineer several mutant forms of the fusogen exhibiting elevated fusion function to improve the overall efficiency of targeted transduction. We devised an animal model to show that subcutaneous injection of such engineered vectors to the areas xenografted with target cells could achieve targeted gene delivery in vivo. Taken together, we demonstrated as proof-of-principle a flexible and modular two-molecule strategy for engineering targeting gamma-retroviral vectors.


Assuntos
Anticorpos/genética , Gammaretrovirus/genética , Marcação de Genes , Técnicas de Transferência de Genes , Vetores Genéticos , Proteínas Recombinantes de Fusão/genética , Animais , Anticorpos/imunologia , Antígenos CD20/genética , Antígenos CD20/imunologia , Linhagem Celular , Feminino , Gammaretrovirus/imunologia , Terapia Genética , Humanos , Camundongos , Proteínas Recombinantes de Fusão/imunologia , Sindbis virus/genética , Transdução Genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
19.
Vaccine ; 24(4): 435-42, 2006 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-16143433

RESUMO

The membrane-proximal external region (MPER) of HIV-1 gp41 is recognized by all three anti-HIV antibodies 2F5, 4E10 and Z13 that were directly derived from AIDS patients and have broader anti-HIV neutralizing activities. Thus, the MPER has been the focus of anti-HIV vaccine design and development. However, it has been unsuccessful to generate anti-HIV neutralizing antibodies targeting this region. One possible reason is that the MPER-containing immunogens have failed to maintain the correct conformation of the MPER present within the HIV-1 viral gp41 protein. The porcine endogenous retrovirus (PERV) p15E protein is structurally similar to HIV-1 gp41, and it was recently reported that the p15E fragment can be expressed in soluble form and was capable of inducing neutralizing antibodies against the epitope within the MPER of PERV p15E. In the present study, we attempted to use the p15E fragment as a carrier and fused the HIV-1 gp41 MPER with the p15E fragment. The vesicular stomatitis virus (VSV) recombinants expressing the fusion proteins (HIV-1 gp41 MPER-p15E) were prepared as primary immunogens, and the soluble fusion protein purified from a baculovirus expression system as booster immunogens. Our results showed that the antisera obtained from immunized rabbits specifically recognized the MPER determinant presented in the gp41 fragment. Importantly, we found that the antisera had neutralizing activities against HIV-1 viruses containing HIV-1 HXB2 and JRFL envelope glycoproteins. These results offer a new strategy for HIV-1 vaccine design and development targeting the gp41 MPER.


Assuntos
Anticorpos Anti-HIV/biossíntese , Proteína gp41 do Envelope de HIV/administração & dosagem , Infecções por HIV/prevenção & controle , HIV-1/imunologia , Vacinas contra a AIDS/administração & dosagem , Vacinas contra a AIDS/imunologia , Animais , Técnicas de Cultura de Células , Gammaretrovirus/genética , Gammaretrovirus/imunologia , Anticorpos Anti-HIV/imunologia , Proteína gp41 do Envelope de HIV/química , Proteína gp41 do Envelope de HIV/genética , Proteína gp41 do Envelope de HIV/imunologia , Infecções por HIV/imunologia , Humanos , Imunização , Testes de Neutralização , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Spodoptera
20.
Xenotransplantation ; 10(6): 562-8, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14708522

RESUMO

Reduction of pig cell-surface alpha-galactosyl (Gal) epitope, Galalpha1, 3Galbeta1, 4GlcNAc-R, by the introduction of glycosyltransferase genes is effective in suppressing hyperacute rejection (HAR) in pig-to-human xenotransplantation. The transmission of porcine endogenous retroviruses (PERVs) has been recognized as a potential risk factor associated with xenotransplantation. In this study, effects of the introduction of glycosyltransferase genes to pig cells on the sensitivity of gammaretroviruses to human serum were investigated. Pig endothelial cells (PEC), PEC transduced with alpha1,2 fucosyltransferase (FT), alpha2,3 sialyltransferase (ST), or N-acetylglucosaminyltransferase III (GnT-III), and human embryonic kidney (HEK) 293 cells were transduced with the LacZ gene with the packaging signal of murine leukemia virus (MuLV) under the control of the long terminal repeat of MuLV by a pseudotype infection. Then, the cells were further infected with PERV subtype B (PERV-B) or feline leukemia virus subgroup B (FeLV-B). Culture supernatants of the infected cells were mixed with human serum (HS) and then inoculated to HEK293 cells. The inoculated cells were histochemically stained and lacZ-positive blue foci were counted. Glycosyltransferase activity, xenoantigenicity, and alpha-Gal epitope density in the cells were measured at the time of the infection experiments. PERV-B or FeLV-B particles from the parental PEC were efficiently neutralized by HS, while those from PEC transduced with alpha1,2FT, alpha2,3ST or GnT-III were less sensitive to HS. The transduced PEC exhibited high levels of activity of the introduced glycotransferases, and expressed fewer xenoantigens and cell-surface alpha-Gal epitopes. Our results suggest that gammaretroviruses including PERVs produced by transgenic pigs, that are generally modified to reduce the cell-surface alpha-Gal epitope to overcome the HAR in xenotransplantation, are less sensitive to HS.


Assuntos
Retrovirus Endógenos/imunologia , Células Endoteliais/virologia , Gammaretrovirus/imunologia , Glicosiltransferases/genética , Soro/imunologia , Animais , Antígenos Heterófilos/imunologia , Linhagem Celular , Retrovirus Endógenos/genética , Células Endoteliais/citologia , Células Endoteliais/enzimologia , Epitopos , Gammaretrovirus/genética , Glicosiltransferases/metabolismo , Humanos , Suínos , Transfecção , Transplante Heterólogo , Zoonoses
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA