Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
PLoS Pathog ; 7(9): e1002265, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21966273

RESUMEN

Natural killer (NK) cells are lymphocytes that play a major role in the elimination of virally-infected cells and tumor cells. NK cells recognize and target abnormal cells through activation of stimulatory receptors such as NKG2D. NKG2D ligands are self-proteins, which are absent or expressed at low levels on healthy cells but are induced upon cellular stress, transformation, or viral infection. The exact molecular mechanisms driving expression of these ligands remain poorly understood. Here we show that murine cytomegalovirus (MCMV) infection activates the phosphatidylinositol-3-kinase (PI3K) pathway and that this activation is required for the induction of the RAE-1 family of mouse NKG2D ligands. Among the multiple PI3K catalytic subunits, inhibition of the p110α catalytic subunit blocks this induction. Similarly, inhibition of p110α PI3K reduces cell surface expression of RAE-1 on transformed cells. Many viruses manipulate the PI3K pathway, and tumors frequently mutate the p110α oncogene. Thus, our findings suggest that dysregulation of the PI3K pathway is an important signal to induce expression of RAE-1, and this may represent a commonality among various types of cellular stresses that result in the induction of NKG2D ligands.


Asunto(s)
Infecciones por Citomegalovirus/fisiopatología , Células Asesinas Naturales/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/biosíntesis , Proteínas Asociadas a Matriz Nuclear/biosíntesis , Proteínas de Transporte Nucleocitoplasmático/biosíntesis , Fosfatidilinositol 3-Quinasa/fisiología , Receptores de Células Asesinas Naturales/fisiología , Animales , Dominio Catalítico/fisiología , Línea Celular Tumoral , Transformación Celular Viral , Fosfatidilinositol 3-Quinasa Clase I , Fibroblastos/virología , Células Asesinas Naturales/inmunología , Ligandos , Ratones , Muromegalovirus/inmunología , Fosfatidilinositol 3-Quinasas/fisiología
2.
J Immunol ; 184(6): 3033-42, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20173027

RESUMEN

The MHC class I (MHC-I) molecules ferry a cargo of peptides to the cell surface as potential ligands for CD8(+) cytotoxic T cells. For nearly 20 years, the cargo has been described as a collection of short 8-9 mer peptides, whose length and sequences were believed to be primarily determined by the peptide-binding groove of MHC-I molecules. Yet the mechanisms for producing peptides of such optimal length and composition have remained unclear. In this study, using mass spectrometry, we determined the amino acid sequences of a large number of naturally processed peptides in mice lacking the endoplasmic reticulum aminopeptidase associated with Ag processing (ERAAP). We find that ERAAP-deficiency changed the oeuvre and caused a marked increase in the length of peptides normally presented by MHC-I. Furthermore, we observed similar changes in the length of viral peptides recognized by CD8(+) T cells in mouse CMV-infected ERAAP-deficient mice. In these mice, a distinct CD8(+) T cell population was elicited with specificity for an N-terminally extended epitope. Thus, the characteristic length, as well as the composition of MHC-I peptide cargo, is determined not only by the MHC-I peptide-binding groove but also by ERAAP proteolysis in the endoplasmic reticulum.


Asunto(s)
Presentación de Antígeno/inmunología , Retículo Endoplásmico/enzimología , Retículo Endoplásmico/inmunología , Antígenos H-2/metabolismo , Leucil Aminopeptidasa/fisiología , Muromegalovirus/inmunología , Fragmentos de Péptidos/metabolismo , Células 3T3 , Secuencia de Aminoácidos , Animales , Presentación de Antígeno/genética , Linfocitos T CD8-positivos/enzimología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Retículo Endoplásmico/virología , Epítopos de Linfocito T/química , Epítopos de Linfocito T/inmunología , Epítopos de Linfocito T/metabolismo , Antígenos H-2/química , Antígenos H-2/inmunología , Antígeno de Histocompatibilidad H-2D , Hibridomas , Hidrólisis , Leucil Aminopeptidasa/deficiencia , Leucil Aminopeptidasa/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Fragmentos de Péptidos/inmunología , Unión Proteica/genética , Unión Proteica/inmunología , Espectrometría de Masas en Tándem , Proteínas Virales/química , Proteínas Virales/inmunología , Proteínas Virales/metabolismo
3.
Nature ; 433(7024): 430-3, 2005 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-15674295

RESUMEN

Endogenous retroviruses are multicopy retroelements accounting for nearly 10% of murine or human genomes. These retroelements spread into our ancestral genome millions of years ago and have acted as a driving force for genome evolution. Endogenous retroviruses may also be deleterious for their host, and have been implicated in cancers and autoimmune diseases. Most retroelements have lost replication competence because of the accumulation of inactivating mutations, but several, including some murine intracisternal A-particle (IAP) and MusD sequences, are still mobile. These elements encode a reverse transcriptase activity and move by retrotransposition, an intracellular copy-and-paste process involving an RNA intermediate. The host has developed mechanisms to silence their expression, mainly cosuppression and gene methylation. Here we identify another level of antiviral control, mediated by APOBEC3G, a member of the cytidine deaminase family that was previously shown to block HIV replication. We show that APOBEC3G markedly inhibits retrotransposition of IAP and MusD elements, and induces G-to-A hypermutations in their DNA copies. APOBEC3G, by editing viral genetic material, provides an ancestral wide cellular defence against endogenous and exogenous invaders.


Asunto(s)
Citidina Desaminasa/metabolismo , Retrovirus Endógenos/genética , Proteínas/metabolismo , Retroelementos/genética , Desaminasa APOBEC-3G , Animales , Secuencia de Bases , Línea Celular , Citidina Desaminasa/genética , Genoma , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Mutagénesis/genética , Nucleósido Desaminasas , Proteínas/genética , Proteínas Represoras , Integración Viral/genética
4.
Curr Cancer Drug Targets ; 20(5): 341-354, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31899676

RESUMEN

Idronoxil has been the subject of more than 50 peer-reviewed publications over the last two decades. This isoflavone is an intriguing regulator of multiple signal transduction pathways, capable of causing a range of biological effects, including cell cycle arrest, apoptosis, an ability to stimulate the immune system, and inhibition of angiogenesis. These multifaceted actions suggest that idronoxil has the potential to synergize with, or complement, a wide range of cancer therapies. Whilst clinically tested in the past, idronoxil's journey was discontinued as a result of its low bioavailability in humans when administered either intravenously or orally, though strategies to overcome this issue are currently being explored. Here, we summarize the current literature regarding the key cellular targets of idronoxil and the mechanisms by which idronoxil exerts its anticancer effects, laying a new foundation toward giving this unique molecule a second chance of contributing to the future of cancer treatment.


Asunto(s)
Antineoplásicos/uso terapéutico , Isoflavonas/uso terapéutico , Neoplasias/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis , Ciclo Celular , Humanos , Neoplasias/metabolismo , Neoplasias/patología
5.
J Virol Methods ; 105(1): 133-40, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12176150

RESUMEN

Human T cell leukemia virus type 1 (HTLV-1) causes two major diseases: adult T-cell leukemia-lymphoma and tropical spastic paraparesis/HTLV-1 associated myelopathy (TSP/HAM). In order to understand the involvement of Tax protein in HTLV-1 pathogenesis, we constructed a HIV-1 based lentiviral vector containing the central DNA flap sequence and either the green fluorescent protein (GFP) or the HTLV-1 tax genes. Using these vectors, GFP and tax genes were introduced in several primary and immortalized cells of endothelial, lymphoid, astrocytic or macrophagic origin. As assessed by GFP expression, up to 100% efficiency of transduction was obtained for all cell types tested. Tax expression was detected by Western blot and immuno-fluorescence in the transduced cells. After transduction, the Tax transcriptional activity was confirmed by the transactivation of HTLV-1 LTR-lacZ or HTLV-1 LTR-GFP reporter genes. Increased CD25 and HLA DR expression was observed in human peripheral blood lymphocytes transduced with the Tax vector. These results indicate that both pathways of Tax transactivation, CREB (viral LTR) and NF-kappa B (CD25 and HLA DR), are functional after transduction by TRIP Tax vector. Therefore, this vector provides a useful tool for investigating the role of the Tax viral protein in the pathogenesis of diseases linked to HTLV-1 infection.


Asunto(s)
Productos del Gen tax/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Lentivirus/genética , Transducción Genética , Línea Celular Transformada , Células Cultivadas , ADN Viral/genética , Productos del Gen tax/metabolismo , Proteínas Fluorescentes Verdes , VIH-1/genética , Infecciones por HTLV-I/fisiopatología , Infecciones por HTLV-I/virología , Virus Linfotrópico T Tipo 1 Humano/metabolismo , Humanos , Operón Lac/genética , Operón Lac/fisiología , Leucocitos Mononucleares/virología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo
6.
Artículo en Inglés | MEDLINE | ID: mdl-26015947

RESUMEN

Gene transfer has therapeutic potential for treating HIV-1 infection by generating cells that are resistant to the virus. We have engineered a novel self-inactivating lentiviral vector, LVsh5/C46, using two viral-entry inhibitors to block early steps of HIV-1 cycle. The LVsh5/C46 vector encodes a short hairpin RNA (shRNA) for downregulation of CCR5, in combination with the HIV-1 fusion inhibitor, C46. We demonstrate here the effective delivery of LVsh5/C46 to human T cell lines, peripheral blood mononuclear cells, primary CD4(+) T lymphocytes, and CD34(+) hematopoietic stem/progenitor cells (HSPC). CCR5-targeted shRNA (sh5) and C46 peptide were stably expressed in the target cells and were able to effectively protect gene-modified cells against infection with CCR5- and CXCR4-tropic strains of HIV-1. LVsh5/C46 treatment was nontoxic as assessed by cell growth and viability, was noninflammatory, and had no adverse effect on HSPC differentiation. LVsh5/C46 could be produced at a scale sufficient for clinical development and resulted in active viral particles with very low mutagenic potential and the absence of replication-competent lentivirus. Based on these in vitro results, plus additional in vivo safety and efficacy data, LVsh5/C46 is now being tested in a phase 1/2 clinical trial for the treatment of HIV-1 disease.

7.
J Exp Med ; 207(10): 2065-72, 2010 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-20819928

RESUMEN

Some mature natural killer (NK) cells cannot be inhibited by major histocompatibility complex (MHC) I molecules, either because they lack corresponding inhibitory receptors or because the host lacks the corresponding MHC I ligands for the receptors. Such NK cells nevertheless remain self-tolerant and exhibit a generalized hyporesponsiveness to stimulation through activating receptors. To address whether NK cell responsiveness is set only during the NK cell differentiation process, we transferred mature NK cells from wild-type (WT) to MHC I-deficient hosts or vice versa. Remarkably, mature responsive NK cells from WT mice became hyporesponsive after transfer to MHC I-deficient mice, whereas mature hyporesponsive NK cells from MHC I-deficient mice became responsive after transfer to WT mice. Altered responsiveness was evident among mature NK cells that had not divided in the recipient animals, indicating that the cells were mature before transfer and that alterations in activity did not require cell division. Furthermore, the percentages of NK cells expressing KLRG1, CD11b, CD27, and Ly49 receptors specific for H-2(b) were not markedly altered after transfer. Thus, the functional activity of mature NK cells can be reset when the cells are exposed to a changed MHC environment. These findings have important implications for how NK cell functions may be curtailed or enhanced in the context of disease.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Animales , Antígeno CD11b/biosíntesis , Antígeno CD11b/inmunología , Diferenciación Celular , División Celular , Antígenos H-2/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Células Asesinas Naturales/trasplante , Lectinas Tipo C , Ratones , Subfamilia A de Receptores Similares a Lectina de Células NK/biosíntesis , Subfamilia A de Receptores Similares a Lectina de Células NK/inmunología , Receptores Inmunológicos/biosíntesis , Receptores Inmunológicos/inmunología , Autotolerancia/inmunología , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
8.
J Virol ; 80(10): 4771-80, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16641270

RESUMEN

Interactions between the oncogenic retrovirus human T-cell leukemia virus type 1 (HTLV-1) and dendritic cells (DCs) are poorly characterized. We show here that monocyte-derived DCs form syncytia and are infected upon coculture with HTLV-1-infected lymphocytes. We examined the role of DC-specific ICAM-3-grabbing nonintegrin (DC-SIGN), a C-type lectin expressed in DCs, in HTLV-1-induced syncytium formation. DC-SIGN is known to bind with high affinity to various viral envelope glycoproteins, including human immunodeficiency virus (HIV) and hepatitis C virus, as well as to the cellular receptors ICAM-2 and ICAM-3. After cocultivating DCs and HTLV-1-infected cells, we found that anti-DC-SIGN monoclonal antibodies (MAbs) were able to decrease the number and size of HTLV-1-induced syncytia. Moreover, expression of the lectin in epithelial-cell lines dramatically enhanced the ability to fuse with HTLV-1-positive cells. Interestingly, in contrast to the envelope (Env) glycoproteins of HIV and other viruses, that of HTLV-1 does not bind directly to DC-SIGN. The facilitating role of the lectin in HTLV-1 syncytium formation is mediated by its interaction with ICAM-2 and ICAM-3, as demonstrated by use of MAbs directed against these adhesion molecules. Altogether, our results indicate that DC-SIGN facilitates HTLV-1 infection and fusion of DCs through an ICAM-dependent mechanism.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Células Dendríticas/fisiología , Virus Linfotrópico T Tipo 1 Humano/fisiología , Lectinas Tipo C/fisiología , Receptores de Superficie Celular/fisiología , Linfocitos T/fisiología , Linfocitos T/virología , Moléculas de Adhesión Celular/metabolismo , Fusión Celular , Línea Celular , Línea Celular Tumoral , Productos del Gen env/metabolismo , Células Gigantes/fisiología , Células Gigantes/virología , Células HeLa , Virus Linfotrópico T Tipo 1 Humano/ultraestructura , Humanos , Molécula 1 de Adhesión Intercelular/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Lectinas Tipo C/metabolismo , Receptores de Superficie Celular/metabolismo
9.
J Virol ; 80(2): 605-14, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16378963

RESUMEN

Foamy viruses (FVs) are nonpathogenic retroviruses infecting many species of mammals, notably primates, cattle, and cats. We have examined whether members of the apolipoprotein B-editing catalytic polypeptide-like subunit (APOBEC) family of antiviral cytidine deaminases restrict replication of simian FV. We show that human APOBEC3G is a potent inhibitor of FV infectivity in cell culture experiments. This antiviral activity is associated with cytidine editing of the viral genome. Both molecular FV clones and primary uncloned viruses were susceptible to APOBEC3G, and viral infectivity was also inhibited by murine and simian APOBEC3G homologues, as well as by human APOBEC3F. Wild-type and bet-deleted viruses were similarly sensitive to this antiviral activity, suggesting that Bet does not significantly counteract APOBEC proteins. Moreover, we did not detect FV sequences that may have been targeted by APOBEC in naturally infected macaques, but we observed a few G-to-A substitutions in humans that have been accidentally contaminated by simian FV. In infected hosts, the persistence strategy employed by FV might be based on low levels of replication, as well as avoidance of cells expressing large amounts of active cytidine deaminases.


Asunto(s)
Citidina Desaminasa/metabolismo , Nucleósido Desaminasas/metabolismo , Proteínas Represoras/metabolismo , Spumavirus/fisiología , Desaminasa APOBEC-3G , Adenina , Animales , Secuencia de Bases , Línea Celular , Citidina Desaminasa/química , Genoma Viral/genética , Guanina , Humanos , Macaca , Ratones , Datos de Secuencia Molecular , Mutación , Especificidad de la Especie , Spumavirus/genética , Replicación Viral
10.
Vaccine ; 23(36): 4463-72, 2005 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-15993518

RESUMEN

Live attenuated measles vaccine (MV) could provide a safe and efficient pediatric vaccination vector to immunize children simultaneously against measles and human immunodeficiency virus type 1 (HIV-1). To evaluate the capacity of a vector derived from the certified Schwarz measles vaccine (MVSchw) to prime effective cytotoxic T cells (CTL) and broad neutralizing antibodies against HIV-1 conserved epitopes, we generated recombinant MVSchw viruses expressing HIV-1 antigens. We demonstrate that a recombinant MVSchw virus expressing an HIV-1-derived CTL polyepitope primes effective HLA-A0201-restricted CTLs against multiple conserved HIV-1 epitopes in mice susceptible to measles and humanized for the major histocompatibility complex (MHC) class-I molecule HLA-A0201. Additionally, we show that a recombinant MVSchw virus expressing an HIV-1(89.6) gp140 glycoprotein whose hyper variable V1, V2 and V3 loops were deleted (DeltaV1V2V3gp140), induces broadly neutralizing antibodies against HIV-1 primary isolates. These results show that the MVSchw pediatric vaccination vector induces efficient cellular and humoral HIV-specific immune responses.


Asunto(s)
Vacunas contra el SIDA/inmunología , Anticuerpos Anti-VIH/sangre , VIH-1/inmunología , Antígenos HLA-A/inmunología , Vacuna Antisarampión/inmunología , Linfocitos T Citotóxicos/inmunología , Vacunas Sintéticas/inmunología , Animales , Antígenos CD/análisis , Recuento de Linfocito CD4 , Epítopos , Productos del Gen env/inmunología , Vectores Genéticos , Inmunización , Interferón-alfa/análisis , Vacuna Antisarampión/genética , Proteína Cofactora de Membrana , Glicoproteínas de Membrana/análisis , Ratones , Vacunas Atenuadas/inmunología , Productos del Gen env del Virus de la Inmunodeficiencia Humana
11.
J Infect Dis ; 191(2): 255-63, 2005 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-15609236

RESUMEN

Human T cell lymphotropic virus (HTLV) type I is the agent of adult T cell leukemia and HTLV-I-associated myelopathy. Because its pathogenesis is not well understood, a mouse model of HTLV-I infection would be valuable. We report the infection of adult BALB/c, C3H/He, 129Sv, and 129Sv IFNAR(-/-) mice with an infectious chimeric HTLV-I provirus bearing the Moloney-murine leukemia virus (Mo-MuLV) envelope glycoprotein truncated for the C-terminal R peptide. Mice were persistently infected (500-800 proviral DNA copies/10(5) splenocytes) for at least 20 weeks after inoculation. The chimeric virus disseminated to several organs, such as spleen, thymus, lung, brain, and spinal cord. The amplification of proviral integration sites showed an oligoclonal integration resembling that reported in HTLV-I-infected humans. Infected mice developed lasting humoral and cellular immune responses. This DeltaR HTLV-I/Mo-MuLV chimeric virus, with the Mo-MuLV Env tropism and HTLV-I replication characteristics, could provide a mouse model of HTLV-I infection.


Asunto(s)
Quimera/inmunología , Virus Linfotrópico T Tipo 1 Humano/inmunología , Virus de la Leucemia Murina de Moloney/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Formación de Anticuerpos/inmunología , Línea Celular , Modelos Animales de Enfermedad , Virus Linfotrópico T Tipo 1 Humano/genética , Humanos , Inmunidad Celular/inmunología , Ratones , Virus de la Leucemia Murina de Moloney/genética , Provirus , Proteínas del Envoltorio Viral/genética
12.
J Gen Virol ; 86(Pt 9): 2489-2494, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16099907

RESUMEN

In the absence of the human immunodeficiency virus type 1 (HIV-1) Vif protein, the host-cell cytidine deaminases APOBEC3F and -3G are co-packaged along with virion RNA. Upon infection of target cells, nascent single-stranded DNA can be edited extensively, invariably giving rise to defective genomes called G-->A hypermutants. Although human T-cell leukemia virus type 1 (HTLV-1) replicates in the same cell type as HIV-1, it was shown here that HTLV-1 is relatively resistant to the antiviral effects mediated by human APOBEC3B, -3C, -3F and -3G. Nonetheless, a small percentage of genomes (0.1

Asunto(s)
Citidina Desaminasa/metabolismo , Genoma Viral , Virus Linfotrópico T Tipo 1 Humano/metabolismo , Edición de ARN , Desaminasa APOBEC-3G , Secuencia de Bases , Citosina Desaminasa/metabolismo , ADN Viral/metabolismo , Virus Linfotrópico T Tipo 1 Humano/química , Virus Linfotrópico T Tipo 1 Humano/genética , Humanos , Leucocitos Mononucleares/virología , Antígenos de Histocompatibilidad Menor , Datos de Secuencia Molecular , Mutación , Nucleósido Desaminasas , Proteínas/metabolismo , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Proteínas Represoras
13.
J Virol ; 76(15): 7883-9, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12097602

RESUMEN

We constructed a chimeric human T-cell lymphotropic virus type 1 (HTLV-1) provirus in which the original envelope precursor sequence was replaced by that of ecotropic Moloney murine leukemia virus (Mo-MuLV). Chimeric particles produced by transient transfection of this chimeric provirus were infectious for murine cells, such as NIH 3T3 fibroblasts, lymphoid EL4 cells, and primary CD4(+) T lymphocytes, whereas HTLV-1 particles were not. The infectivity of chimeric particles increased 10 times when the R peptide located at the carboxy terminus of the MuLV envelope glycoprotein was deleted. Primary murine CD4(+) T lymphocytes, infected by the Delta R chimeric virus, released particles that could spread the infection to other naive murine lymphoid cells. This chimeric virus, with the Mo-MuLV envelope glycoprotein and the replication characteristics of HTLV-1, should be useful in studying the pathogenesis of HTLV-1 in a mouse model.


Asunto(s)
Virus Linfotrópico T Tipo 1 Humano/genética , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Virus de la Leucemia Murina de Moloney/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteínas del Envoltorio Viral/genética , Células 3T3 , Secuencia de Aminoácidos , Animales , Linfocitos T CD4-Positivos/virología , Virus Linfotrópico T Tipo 1 Humano/metabolismo , Humanos , Linfocitos , Ratones , Datos de Secuencia Molecular , Virus de la Leucemia Murina de Moloney/genética , Provirus , Proteínas Recombinantes de Fusión/genética , Transfección , Proteínas del Envoltorio Viral/metabolismo
14.
J Virol ; 78(1): 146-57, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14671096

RESUMEN

The anchored and secreted forms of the human immunodeficiency virus type 1 (HIV-1) 89.6 envelope glycoprotein, either complete or after deletion of the V3 loop, were expressed in a cloned attenuated measles virus (MV) vector. The recombinant viruses grew as efficiently as the parental virus and expressed high levels of the HIV protein. Expression was stable during serial passages. The immunogenicity of these recombinant vectors was tested in mice susceptible to MV and in macaques. High titers of antibodies to both MV and HIV-Env were obtained after a single injection in susceptible mice. These antibodies neutralized homologous SHIV89.6p virus, as well as several heterologous HIV-1 primary isolates. A gp160 mutant in which the V3 loop was deleted induced antibodies that neutralized heterologous viruses more efficiently than antibodies induced by the native envelope protein. A high level of CD8+ and CD4+ cells specific for HIV gp120 was also detected in MV-susceptible mice. Furthermore, recombinant MV was able to raise immune responses against HIV in mice and macaques with a preexisting anti-MV immunity. Therefore, recombinant MV vaccines inducing anti-HIV neutralizing antibodies and specific T lymphocytes responses deserve to be tested as a candidate AIDS vaccine.


Asunto(s)
Vacunas contra el SIDA/administración & dosificación , Productos del Gen env/inmunología , Anticuerpos Anti-VIH/sangre , Virus del Sarampión/genética , Linfocitos T/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas contra el SIDA/genética , Vacunas contra el SIDA/inmunología , Animales , Reacciones Cruzadas , Productos del Gen env/administración & dosificación , Productos del Gen env/genética , Infecciones por VIH/prevención & control , Infecciones por VIH/virología , Humanos , Inmunización , Macaca , Virus del Sarampión/inmunología , Ratones , Pruebas de Neutralización , Vacunas Sintéticas/inmunología
15.
J Virol ; 77(21): 11546-54, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14557640

RESUMEN

Live attenuated RNA viruses make highly efficient vaccines. Among them, measles virus (MV) vaccine has been given to a very large number of children and has been shown to be highly efficacious and safe. Therefore, this vaccine might be a very promising vector to immunize children against both measles and other infectious agents, such as human immunodeficiency virus. A vector was previously derived from the Edmonston B strain of MV, a vaccine strain abandoned 25 years ago. Sequence analysis revealed that the genome of this vector diverges from Edmonston B by 10 amino acid substitutions not related to any Edmonston subgroup. Here we describe an infectious cDNA for the Schwarz/Moraten strain, a widely used MV vaccine. This cDNA was constructed from a batch of commercial vaccine. The extremities of the cDNA were engineered in order to maximize virus yield during rescue. A previously described helper cell-based rescue system was adapted by cocultivating transfected cells on primary chicken embryo fibroblasts, the cells used to produce the Schwarz/Moraten vaccine. After two passages the sequence of the rescued virus was identical to that of the cDNA and of the published Schwarz/Moraten sequence. Two additional transcription units were introduced in the cDNA for cloning foreign genetic material. The immunogenicity of rescued virus was studied in macaques and in mice transgenic for the CD46 MV receptor. Antibody titers and T-cell responses (ELISpot) in animals inoculated with low doses of rescued virus were identical to those obtained with commercial Schwarz MV vaccine. In contrast, the immunogenicity of the previously described Edmonston B strain-derived MV clone was much lower. This new molecular clone will allow for the production of MV vaccine without having to rely on seed stocks. The additional transcription units allow expressing heterologous antigens, thereby providing polyvalent vaccines based on an approved, safe, and efficient MV vaccine strain that is used worldwide.


Asunto(s)
Anticuerpos Antivirales/sangre , Clonación Molecular , Vacuna Antisarampión/inmunología , Virus del Sarampión/genética , Secuencia de Aminoácidos , Animales , Antígenos CD/genética , Secuencia de Bases , Embrión de Pollo , Chlorocebus aethiops , ADN Complementario/genética , Humanos , Inmunización , Macaca , Sarampión/prevención & control , Sarampión/virología , Vacuna Antisarampión/genética , Virus del Sarampión/clasificación , Proteína Cofactora de Membrana , Glicoproteínas de Membrana/genética , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Recombinación Genética , Análisis de Secuencia de ADN , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA