Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Gene Ther ; 26(5): 151-164, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30718755

RESUMEN

Induced pluripotent stem cells (iPSCs) provide a unique platform for individualized cell therapy approaches. Currently, episomal DNA, mRNA, and Sendai virus-based RNA reprogramming systems are widely used to generate iPSCs. However, they all rely on the use of multiple (three to six) components (vectors/plasmids/mRNAs) leading to the production of partially reprogrammed cells, reducing the efficiency of the systems. We produced a one-cycle measles virus (MV) vector by substituting the viral attachment protein gene with the green fluorescent protein (GFP) gene. Here, we present a highly efficient multi-transgene delivery system based on a vaccine strain of MV, a non-integrating RNA virus that has a long-standing safety record in humans. Introduction of the four reprogramming factors OCT4, SOX2, KLF4, and cMYC via a single, "one-cycle" MV vector efficiently reprogrammed human somatic cells into iPSCs, whereas MV vector genomes are rapidly eliminated in derived iPSCs. Our MV vector system offers a new reprogramming platform for genomic modification-free iPSCs amenable for clinical translation.


Asunto(s)
Técnicas de Reprogramación Celular/métodos , Reprogramación Celular , Técnicas de Transferencia de Gen , Células Madre Pluripotentes Inducidas/citología , Virus del Sarampión/genética , Animales , Células Cultivadas , Chlorocebus aethiops , Vectores Genéticos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Células Vero
2.
J Virol ; 88(18): 10851-63, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25008930

RESUMEN

UNLABELLED: The genome of nonsegmented negative-strand RNA viruses is tightly embedded within a nucleocapsid made of a nucleoprotein (N) homopolymer. To ensure processive RNA synthesis, the viral polymerase L in complex with its cofactor phosphoprotein (P) binds the nucleocapsid that constitutes the functional template. Measles virus P and N interact through two binding sites. While binding of the P amino terminus with the core of N (NCORE) prevents illegitimate encapsidation of cellular RNA, the interaction between their C-terminal domains, P(XD) and N(TAIL) is required for viral RNA synthesis. To investigate the binding dynamics between the two latter domains, the P(XD) F497 residue that makes multiple hydrophobic intramolecular interactions was mutated. Using a quantitative mammalian protein complementation assay and recombinant viruses, we found that an increase in P(XD)-to-N(TAIL) binding strength is associated with a slower transcript accumulation rate and that abolishing the interaction renders the polymerase nonfunctional. The use of a newly developed system allowing conditional expression of wild-type or mutated P genes, revealed that the loss of the P(XD)-N(TAIL) interaction results in reduced transcription by preformed transcriptases, suggesting reduced engagement on the genomic template. These intracellular data indicate that the viral polymerase entry into and progression along its genomic template relies on a protein-protein interaction that serves as a tightly controlled dynamic anchor. IMPORTANCE: Mononegavirales have a unique machinery to replicate RNA. Processivity of their polymerase is only achieved when the genome template is entirely embedded into a helical homopolymer of nucleoproteins that constitutes the nucleocapsid. The polymerase binds to the nucleocapsid template through the phosphoprotein. How the polymerase complex enters and travels along the nucleocapsid template to ensure uninterrupted synthesis of up to ∼ 6,700-nucleotide messenger RNAs from six to ten consecutive genes is unknown. Using a quantitative protein complementation assay and a biGene-biSilencing system allowing conditional expression of two P genes copies, the role of the P-to-N interaction in polymerase function was further characterized. We report here a dynamic protein anchoring mechanism that differs from all other known polymerases that rely only onto a sustained and direct binding to their nucleic acid template.


Asunto(s)
Virus del Sarampión/fisiología , Sarampión/virología , Nucleocápside/metabolismo , Fosfoproteínas/metabolismo , Proteínas Virales/metabolismo , Replicación Viral , Regulación Viral de la Expresión Génica , Humanos , Virus del Sarampión/química , Virus del Sarampión/genética , Nucleocápside/química , Nucleocápside/genética , Fosfoproteínas/química , Fosfoproteínas/genética , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Virales/química , Proteínas Virales/genética
3.
bioRxiv ; 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38798498

RESUMEN

DddA-derived cytosine base editors (DdCBEs) enable the targeted introduction of C•G-to-T•A conversions in mitochondrial DNA (mtDNA). DdCBEs are often deployed as pairs, with each arm comprised of a transcription activator-like effector (TALE), a split double-stranded DNA deaminase half, and a uracil glycosylase inhibitor. This pioneering technology has helped improve our understanding of cellular processes involving mtDNA and has paved the way for the development of models and therapies for genetic disorders caused by pathogenic mtDNA variants. Nonetheless, given the intrinsic properties of TALE proteins, several target sites in human mtDNA remain out of reach to DdCBEs and other TALE-based technologies. Specifically, due to the conventional requirement for a thymine immediately upstream of the TALE target sequences (i.e., the 5'-T constraint), over 150 loci in the human mitochondrial genome are presumed to be inaccessible to DdCBEs. Previous attempts at circumventing this constraint, either by developing monomeric DdCBEs or utilizing DNA-binding domains alternative to TALEs, have resulted in suboptimal specificity profiles with reduced therapeutic potential. Here, aiming to challenge and elucidate the relevance of the 5'-T constraint in the context of DdCBE-mediated mtDNA editing, and to expand the range of motifs that are editable by this technology, we generated αDdCBEs that contain modified TALE proteins engineered to recognize all 5' bases. Notably, 5'-T-noncompliant, canonical DdCBEs efficiently edited mtDNA at diverse loci. However, DdCBEs were frequently outperformed by αDdCBEs, which consistently displayed significant improvements in activity and specificity, regardless of the 5'-most bases of their TALE binding sites. Furthermore, we showed that αDdCBEs are compatible with DddA tox and its derivatives DddA6, and DddA11, and we validated TALE shifting with αDdCBEs as an effective approach to optimize base editing outcomes at a single target site. Overall, αDdCBEs enable efficient, specific, and unconstrained mitochondrial base editing.

4.
J Virol ; 85(1): 348-56, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20980517

RESUMEN

Measles remains a leading cause of death worldwide among children because it suppresses immune function. The measles virus (MV) P gene encodes three proteins (P, V, and C) that interfere with innate immunity, controlling STAT1, STAT2, mda5, and perhaps other key regulators of immune function. We identified here three residues in the shared domain of the P and V proteins-tyrosine 110, valine 112, and histidine 115-that function to retain STAT1 in the cytoplasm and inhibit interferon transcription. This information was used to generate a recombinant measles virus unable to antagonize STAT1 function (STAT1-blind MV) differing only in these three residues from a wild-type strain of well-defined virulence. This virus was used to assess the relevance of P and V interactions with STAT1 for virulence in primates. When a group of six rhesus monkeys (Macaca mulatta) was inoculated intranasally with STAT1-blind MV, viremia was short-lived, and the skin rash and other clinical signs observed with wild-type MV were absent. The STAT1-blind virus less efficiently controlled the inflammatory response, as measured by enhanced transcription of interleukin-6 and tumor necrosis factor alpha in peripheral blood mononuclear cells from infected hosts. Importantly, neutralizing antibody titers and MV-specific T-cell responses were equivalent in hosts infected with either virus. These findings indicate that efficient MV interactions with STAT1 are required to sustain virulence in a natural host by controlling the inflammatory response against the virus. They also suggest that selectively STAT1-blind MV may have utility as vectors for targeted oncolysis and vaccination.


Asunto(s)
Inflamación/prevención & control , Virus del Sarampión/patogenicidad , Fosfoproteínas/genética , Recombinación Genética , Factor de Transcripción STAT1/antagonistas & inhibidores , Proteínas Virales/genética , Secuencia de Aminoácidos , Animales , Chlorocebus aethiops , Células HeLa , Humanos , Inflamación/inmunología , Macaca mulatta , Sarampión/inmunología , Sarampión/prevención & control , Sarampión/virología , Vacuna Antisarampión/genética , Vacuna Antisarampión/inmunología , Virus del Sarampión/genética , Datos de Secuencia Molecular , Mutación , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Células Vero , Proteínas Virales/química , Proteínas Virales/metabolismo , Virulencia
5.
Mol Diagn Ther ; 26(4): 353-367, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35763161

RESUMEN

Ethical issues are a significant barrier to the use of embryonic stem cells in patients due to their origin: human embryos. To further the development of stem cells in a patient application, alternative sources of cells were sought. A process referred to as reprogramming was established to create induced pluripotent stem cells from somatic cells, resolving the ethical issues, and vectors were developed to deliver the reprogramming factors to generate induced pluripotent stem cells. Early viral vectors used integrating retroviruses and lentiviruses as delivery vehicles for the transcription factors required to initiate reprogramming. However, because of the inherent risk associated with vectors that integrate into the host genome, non-integrating approaches were explored. The development of non-integrating viral vectors offers a safer alternative, and these modern vectors are reliable, efficient, and easy to use to achieve induced pluripotent stem cells suitable for direct patient application in the growing field of individualized medicine. This review summarizes all the RNA viral vectors in the field of reprogramming with a special focus on the emerging delivery vectors based on non-integrating  Paramyxoviruses, Sendai and measles viruses. We discuss their design and evolution towards being safe and efficient reprogramming vectors in generating induced pluripotent stem cells from somatic cells.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas , Reprogramación Celular/genética , Vectores Genéticos/genética , Humanos , ARN , Tecnología
6.
Mol Ther Methods Clin Dev ; 24: 48-61, 2022 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-34977272

RESUMEN

OCT4 is a key mediator of induced pluripotent stem cell (iPSC) reprogramming, but the mechanistic insights into the role of exogenous OCT4 and timelines that initiate pluripotency remain to be resolved. Here, using measles reprogramming vectors, we present microRNA (miRNA) targeting of exogenous OCT4 to shut down its expression during the mesenchymal to the epithelial transition phase of reprogramming. We showed that exogenous OCT4 is required only for the initiation of reprogramming and is dispensable for the maturation stage. However, the continuous expression of SOX2, KLF4, and c-MYC is necessary for the maturation stage of the iPSC. Additionally, we demonstrate a novel application of miRNA targeting in a viral vector to contextually control the vector/transgene, ultimately leading to an improved reprogramming efficiency. This novel approach could be applied to other systems for improving the efficiency of vector-induced processes.

7.
J Clin Invest ; 118(7): 2448-58, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18568079

RESUMEN

The current model of measles virus (MV) pathogenesis implies that apical infection of airway epithelial cells precedes systemic spread. An alternative model suggests that primarily infected lymphatic cells carry MV to the basolateral surface of epithelial cells, supporting MV shedding into the airway lumen and contagion. This model predicts that a mutant MV, unable to enter cells through the unidentified epithelial cell receptor (EpR), would remain virulent but not be shed. To test this model, we identified residues of the MV attachment protein sustaining EpR-mediated cell fusion. These nonpolar or uncharged polar residues defined an area located near the binding site of the signaling lymphocytic activation molecule (SLAM), the receptor for MV on lymphatic cells. We then generated an EpR-blind virus maintaining SLAM-dependent cell entry and inoculated rhesus monkeys intranasally. Hosts infected with the selectively EpR-blind MV developed rash and anorexia while averaging slightly lower viremia than hosts infected with wild-type MV but did not shed virus in the airways. The mechanism restricting shedding was characterized using primary well-differentiated human airway epithelial cells. Wild-type MV infected columnar epithelial cells bearing tight junctions only when applied basolaterally, while the EpR-blind virus did not infect these cells. Thus, EpR is probably a basolateral protein, and infection of the airway epithelium is not essential for systemic spread and virulence of MV.


Asunto(s)
Virus del Sarampión/patogenicidad , Sarampión/metabolismo , Receptores Virales/metabolismo , Proteínas Virales/metabolismo , Esparcimiento de Virus/fisiología , Secuencia de Aminoácidos , Animales , Formación de Anticuerpos/inmunología , Antígenos CD/metabolismo , Línea Celular Transformada , Línea Celular Tumoral , Células Epiteliales/metabolismo , Células Epiteliales/virología , Femenino , Haplorrinos , Humanos , Leucocitos Mononucleares/virología , Macaca mulatta , Masculino , Sarampión/transmisión , Sarampión/virología , Virus del Sarampión/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Receptores de Superficie Celular/metabolismo , Mucosa Respiratoria/virología , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Proteínas Virales/química , Proteínas Virales/genética , Virulencia , Acoplamiento Viral
8.
J Virol ; 84(1): 380-6, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19846517

RESUMEN

The measles virus P gene products V and C antagonize the host interferon (IFN) response, blocking both IFN signaling and production. Using Moraten vaccine strain-derived measles virus and isogenic mutants deficient for either V or C protein production (V(ko) and C(ko), respectively), we observed that the C(ko) virus was a potent inducer of IFN-beta, while induction by V(ko) virus was an order of magnitude lower than that by the C(ko) virus. The parental recombinant Moraten virus did not significantly induce IFN-beta. The enhanced IFN-inducing capacity of the C(ko) virus correlated with an enhanced activation of IFN regulatory factor 3 (IRF-3), NF-kappaB, and ATF-2 in C(ko)-infected compared to V(ko) or parental virus-infected cells. Furthermore, protein kinase PKR and mitochondrial adapter IPS-1 were required for maximal C(ko)-mediated IFN-beta induction, which correlated with the PKR-mediated enhancement of mitogen-activated protein kinase and NF-kappaB activation. Our results reveal multiple consequences of C protein expression and document an important function for PKR as an enhancer of IFN-beta induction during measles virus infection.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Interferón beta/genética , Virus del Sarampión/inmunología , Proteínas Virales/fisiología , eIF-2 Quinasa/fisiología , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Chlorocebus aethiops , Células HeLa , Humanos , Virus del Sarampión/química , FN-kappa B/metabolismo , Células Vero
9.
J Virol ; 83(2): 961-8, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19004947

RESUMEN

The measles virus (MV) accessory proteins V and C play important roles in MV replication and pathogenesis. Infection with recombinant MV lacking either V or C causes more cell death than infection with the parental vaccine-equivalent virus (MVvac), and C-deficient virus grows poorly relative to the parental virus. Here, we show that a major effector of the C phenotype is the RNA-dependent protein kinase PKR. Using human HeLa cells stably deficient in PKR as a result of RNA interference-mediated knockdown (PKR(kd) cells), we demonstrated that a reduction in PKR partially rescued the growth defect of C knockout (C(ko)) virus but had no effect on the growth of either wild-type (WT) or V knockout (V(ko)) virus. Increased growth of the C(ko) virus in PKR(kd) cells correlated with increased viral protein expression, while defective growth and decreased protein expression in PKR-sufficient cells correlated with increased phosphorylation of PKR and the alpha subunit of eukaryotic initiation factor 2. Furthermore, infection with WT, V(ko), or especially C(ko) virus caused significantly less apoptosis in PKR(kd) cells than in PKR-sufficient cells. Although apoptosis induced by C(ko) virus infection in PKR-sufficient cells was blocked by a caspase antagonist, the growth of C(ko) virus was not restored to the WT level by treatment with this pharmacologic inhibitor. Taken together, these results indicate that PKR plays an important antiviral role during MV infection but that the virus growth restriction by PKR is not dependent upon the induction of apoptosis. Furthermore, the results establish that a principal function of the MV C protein is to antagonize the proapoptotic and antiviral activities of PKR.


Asunto(s)
Apoptosis , Virus del Sarampión/crecimiento & desarrollo , Virus del Sarampión/inmunología , Proteínas no Estructurales Virales/genética , eIF-2 Quinasa/inmunología , Animales , Chlorocebus aethiops , Factor 2 Eucariótico de Iniciación , Eliminación de Gen , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Células HeLa , Humanos , Virus del Sarampión/genética , Fosforilación , Células Vero , Proteínas no Estructurales Virales/biosíntesis , Proteínas no Estructurales Virales/fisiología , eIF-2 Quinasa/metabolismo
10.
J Virol ; 82(11): 5359-67, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18385234

RESUMEN

Patients recruited in virus-based cancer clinical trials and immunocompromised individuals in need of vaccination would profit from viral strains with defined attenuation mechanisms. We generated measles virus (MV) strains defective for the expression of either the V protein, a modulator of the innate immune response, or the C protein, which has multiple functions. The virulence of these strains was compared with that of the parental wild-type MV in a natural host, Macaca mulatta. Skin rash, viremia, and the strength of the innate and adaptive immune responses were characterized in groups of six animals. Replication of V- or C-protein-defective viruses was short-lived and reached lower levels in peripheral blood mononuclear cells and lymphatic organs compared to the wild-type virus; none of the mutants reverted to the wild type. The neutralizing antibody titers and MV-specific T-cell responses were equivalent in monkeys infected with the viral strains tested, documenting strong adaptive immune responses. In contrast, the inflammatory response was better controlled by wild-type MV, as revealed by inhibition of interleukin-6 and tumor necrosis factor alpha transcription. The interferon response was also better controlled by the wild-type virus than by the defective viruses. Since V- and C-defective MVs induce strong adaptive immune responses while spreading less efficiently, they may be developed as vaccines for immunocompromised individuals. Moreover, MV unable to interact with single innate immunity proteins may be developed for preferential replication in tumors with specific contexts of vulnerability.


Asunto(s)
Control de Infecciones , Interferones/biosíntesis , Macaca mulatta/inmunología , Virus del Sarampión/inmunología , Virus del Sarampión/metabolismo , Proteínas Virales/inmunología , Proteínas Virales/metabolismo , Adaptación Fisiológica/inmunología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Chlorocebus aethiops , Humanos , Inflamación/prevención & control , Interferones/genética , Sarampión/genética , Sarampión/inmunología , Sarampión/metabolismo , Sarampión/transmisión , Virus del Sarampión/química , Virus del Sarampión/genética , Datos de Secuencia Molecular , Mutación/genética , Células TH1/inmunología , Células Th2/inmunología , Proteínas Virales/química , Proteínas Virales/genética , Replicación Viral
11.
Stem Cell Res Ther ; 6: 48, 2015 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-25889591

RESUMEN

INTRODUCTION: Advances in the field of stem cells have led to novel avenues for generating induced pluripotent stem cells (iPSCs) from differentiated somatic cells. iPSCs are typically obtained by the introduction of four factors--OCT4, SOX2, KLF4, and cMYC--via integrating vectors. Here, we report the feasibility of a novel reprogramming process based on vectors derived from the non-integrating vaccine strain of measles virus (MV). METHODS: We produced a one-cycle MV vector by substituting the viral attachment protein gene with the green fluorescent protein (GFP) gene. This vector was further engineered to encode for OCT4 in an additional transcription unit. RESULTS: After verification of OCT4 expression, we assessed the ability of iPSC reprogramming. The reprogramming vector cocktail with the OCT4-expressing MV vector and SOX2-, KLF4-, and cMYC-expressing lentiviral vectors efficiently transduced human skin fibroblasts and formed iPSC colonies. Reverse transcription-polymerase chain reaction and immunostaining confirmed induction of endogenous pluripotency-associated marker genes, such as SSEA-4, TRA-1-60, and Nanog. Pluripotency of derived clones was confirmed by spontaneous differentiation into three germ layers, teratoma formation, and guided differentiation into beating cardiomyocytes. CONCLUSIONS: MV vectors can induce efficient nuclear reprogramming. Given the excellent safety record of MV vaccines and the translational capabilities recently developed to produce MV-based vectors now used for cancer clinical trials, our MV vector system provides an RNA-based, non-integrating gene transfer platform for nuclear reprogramming that is amenable for immediate clinical translation.


Asunto(s)
Reprogramación Celular/genética , Células Madre Pluripotentes Inducidas/citología , Virus del Sarampión/genética , Miocitos Cardíacos/citología , Factor 3 de Transcripción de Unión a Octámeros/genética , Animales , Antígenos de Superficie/genética , Biomarcadores , Línea Celular , Reprogramación Celular/fisiología , Chlorocebus aethiops , Fibroblastos/citología , Prepucio/citología , Células HEK293 , Proteínas de Homeodominio/genética , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/biosíntesis , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Ratones , Ratones SCID , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Proteoglicanos/genética , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Proteínas Proto-Oncogénicas c-myc/genética , Factores de Transcripción SOXB1/biosíntesis , Factores de Transcripción SOXB1/genética , Piel/citología , Antígenos Embrionarios Específico de Estadio/genética , Células Vero
12.
Virology ; 444(1-2): 250-6, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23856440

RESUMEN

The measles virus (MV) phosphoprotein (P) and V proteins block the interferon (IFN) response by impeding phosphorylation of the signal transducer and activator of transcription 1 (STAT1) by the Janus kinase 1 (JAK1). We characterized how STAT1 mutants interact with P and JAK1 phosphorylation. Certain mutants of the linker, the Src-homology 2 domain (SH2), or the transactivation domain had reduced or abolished phosphorylation through JAK1 after IFN treatment. Other mutants, mainly localized in the linker, failed to interact with P as documented by the lack of interference with nuclear translocation. Thus the functional footprint of P on STAT1 localizes mainly to the linker domain; there is also some overlap with the STAT1 phosphorylation functional footprint on the SH2 domain. Based on these observations, we discuss how the MV-P might operate to inhibit the JAK/STAT pathway.


Asunto(s)
Interacciones Huésped-Patógeno , Virus del Sarampión/fisiología , Fosfoproteínas/metabolismo , Dominios y Motivos de Interacción de Proteínas , Factor de Transcripción STAT1/metabolismo , Proteínas Virales/metabolismo , Línea Celular , Análisis Mutacional de ADN , Humanos , Mapeo de Interacción de Proteínas , Factor de Transcripción STAT1/genética
13.
J Infect Dis ; 196(4): 541-9, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17624839

RESUMEN

The mechanisms of measles virus (MV) vaccine attenuation are insufficiently characterized. Because the Edmonston vaccine strain can enter cells through CD46 in addition to the primary MV receptor signaling lymphocyte activation molecule (SLAM or CD150), we asked whether and how its tropism is altered. In human tonsillar tissue, this vaccine strain infects naive (CD45RA(+)CD62L(+)) T lymphocytes, which express SLAM very infrequently, with much higher efficiency than do wild-type strains. By contrast, it infects B lymphocytes, macrophages, and NK cells with significantly lower efficiencies than those of wild-type strains. Infection levels by wild-type strains correlate with the frequency of SLAM expression and are highest in B cells, which are 40%-55% infected. SLAM-expressing T cells are more readily infected by all MV strains than are SLAM-expressing B cells. Thus, vaccine attenuation may be caused by tropism alteration in combination with suboptimal replication.


Asunto(s)
Vacuna Antisarampión/metabolismo , Virus del Sarampión/fisiología , Sarampión/virología , Animales , Antígenos CD/metabolismo , Linfocitos B/metabolismo , Linfocitos B/virología , Callithrix , Células Cultivadas , Chlorocebus aethiops , Humanos , Memoria Inmunológica , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/virología , Macrófagos/metabolismo , Macrófagos/virología , Proteína Cofactora de Membrana/metabolismo , Tonsila Palatina/metabolismo , Tonsila Palatina/virología , Receptores de Superficie Celular/metabolismo , Receptores Virales/metabolismo , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/virología , Vacunas Atenuadas/metabolismo , Células Vero , Replicación Viral
14.
J Virol ; 81(19): 10597-605, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17634218

RESUMEN

Hepatitis B virus (HBV) acute and chronic infections remain a major worldwide health problem. Towards developing an anti-HBV vaccine with single-dose scheme potential, we engineered infectious measles virus (MV) genomic cDNAs with a vaccine strain background and expression vector properties. Hepatitis B surface antigen (HBsAg) expression cassettes were inserted into this cDNA and three MVs expressing HBsAg at different levels generated. All vectored MVs, which secrete HBsAg as subviral particles, elicited humoral responses in MV-susceptible genetically modified mice. However, small differences in HBsAg expression elicited vastly different HBsAg antibody levels. The two vectors inducing the highest HBsAg antibody levels were inoculated into rhesus monkeys (Macaca mulatta). After challenge with a pathogenic MV strain (Davis87), control naive monkeys showed a classic measles rash and high viral loads. In contrast, all monkeys immunized with vaccine or a control nonvectored recombinant vaccine or HBsAg-expressing vectored MV remained healthy, with low or undetectable viral loads. After a single vaccine dose, only the vector expressing HBsAg at the highest levels elicited protective levels of HBsAg antibodies in two of four animals. These observations reveal an expression threshold for efficient induction of HBsAg humoral immune responses. This threshold is lower in mice than in macaques. Implications for the development of divalent vaccines based on live attenuated viruses are discussed.


Asunto(s)
Anticuerpos contra la Hepatitis B/inmunología , Antígenos de Superficie de la Hepatitis B/inmunología , Vacunas contra Hepatitis B/inmunología , Hepatitis B/prevención & control , Virus del Sarampión/genética , Sarampión/prevención & control , Animales , Formación de Anticuerpos , Secuencia de Bases , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Anticuerpos contra la Hepatitis B/sangre , Antígenos de Superficie de la Hepatitis B/genética , Vacunas contra Hepatitis B/genética , Macaca mulatta , Virus del Sarampión/inmunología , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular
15.
Virology ; 360(1): 72-83, 2007 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-17112561

RESUMEN

The measles virus (MV) P gene encodes three proteins: P, an essential polymerase cofactor, and C and V, which have multiple functions including immune evasion. We show here that the MV P protein also contributes to immune evasion, and that tyrosine 110 is required to block nuclear translocation of the signal transducer and activator of transcription factors (STAT) after interferon type I treatment. In particular, MV P inhibits STAT1 phosphorylation. This is shown not only by transient expression but also by reverse genetic analyses based on a new functional infectious cDNA derived from a MV vaccine vial (Moraten strain). Our study also identifies a conserved sequence around P protein tyrosine 110 as a candidate interaction site with a cellular protein.


Asunto(s)
Virus del Sarampión/metabolismo , Sarampión/virología , Fosfoproteínas/metabolismo , Factor de Transcripción STAT1/metabolismo , Tirosina/fisiología , Proteínas Virales/metabolismo , Animales , Secuencia de Bases , Núcleo Celular/metabolismo , Chlorocebus aethiops , Células HeLa , Humanos , Inmunidad Innata , Interferón Tipo I/farmacología , Sarampión/inmunología , Datos de Secuencia Molecular , Fosfoproteínas/química , Fosfoproteínas/genética , Fosforilación/efectos de los fármacos , Alineación de Secuencia , Transducción de Señal , Células Vero , Proteínas Virales/química , Proteínas Virales/genética
16.
J Virol ; 80(11): 5644-50, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16699046

RESUMEN

Paramyxovirus V proteins function as host interference factors that inactivate antiviral responses, including interferon. Characterization of cellular proteins that copurify with ectopically expressed measles virus V protein has revealed interactions with DNA binding domains of p53 family proteins, p53 and p73. Specific transcriptional assays reveal that expression of measles virus V cDNA inhibits p73, but not p53. Expression of measles virus V cDNA can delay cell death induced by genotoxic stress and also can decrease the abundance of the proapoptotic factor PUMA, a p73 target. Recombinant measles virus with an engineered deficiency in V protein is capable of inducing more severe cytopathic effects than the wild type, implicating measles virus V protein as an inhibitor of cell death. These findings also suggest that p73-PUMA signaling may be a previously unrecognized arm of cellular innate antiviral immunity.


Asunto(s)
Proteínas de Unión al ADN/antagonistas & inhibidores , Virus del Sarampión/química , Proteínas Nucleares/antagonistas & inhibidores , Fosfoproteínas/farmacología , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Virales/farmacología , Proteínas de Unión al ADN/metabolismo , Humanos , Proteínas Nucleares/metabolismo , Linfocitos T/virología , Proteína Tumoral p73 , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo
17.
J Virol ; 78(21): 11632-40, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15479804

RESUMEN

The measles virus (MV) P gene codes for three proteins: P, an essential polymerase cofactor, and V and C, which have multiple functions but are not strictly required for viral propagation in cultured cells. V shares the amino-terminal domain with P but has a zinc-binding carboxyl-terminal domain, whereas C is translated from an overlapping reading frame. During replication, the P protein binds incoming monomeric nucleocapsid (N) proteins with its amino-terminal domain and positions them for assembly into the nascent ribonucleocapsid. The P protein amino-terminal domain is natively unfolded; to probe its conformational flexibility, we fused it to the green fluorescent protein (GFP), thereby also silencing C protein expression. A recombinant virus (MV-GFP/P) expressing hybrid GFP/P and GFP/V proteins in place of standard P and V proteins and not expressing the C protein was rescued and produced normal ratios of mono-, bi-, and tricistronic RNAs, but its replication was slower than that of the parental virus. Thus, the P protein retained nearly intact polymerase cofactor function, even with a large domain added to its amino terminus. Having noted that titers of cell-associated and especially released MV-GFP/P were reduced and knowing that the C protein of the related Sendai virus has particle assembly and infectivity factor functions, we produced an MV-GFP/P derivative expressing C. Intracellular titers of this virus were almost completely restored, and those of released virus were partially restored. Thus, the MV C protein is an infectivity factor.


Asunto(s)
Virus del Sarampión/patogenicidad , Fosfoproteínas/química , Proteínas Virales/química , Proteínas Virales/fisiología , Animales , Chlorocebus aethiops , Conformación Proteica , Células Vero , Replicación Viral
18.
J Gen Virol ; 85(Pt 6): 1665-1673, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15166451

RESUMEN

Measles virus (MV)-infected cells are activators of the alternative human complement pathway, resulting in high deposition of C3b on the cell surface. Activation was observed independent of whether CD46 was used as a cellular receptor and did not correlate with CD46 down-regulation. The virus itself was an activator of the alternative pathway and was covered by C3b/C3bi, resulting in some loss in infectivity without loss of virus binding to target cells. The cell surface expression of MV fusion (F), but not haemagglutinin, envelope protein resulted in complement activation of the Factor B-dependent alternative pathway in a dose-dependent manner and F-C3b complexes were formed. The underlying activation mechanism was not related to any decrease in cell surface expression of the complement regulators CD46 and CD55. The C3b/C3bi coating of MV-infected cells and virus should ensure enhanced targeting of MV antigens to the immune system, through binding to complement receptors.


Asunto(s)
Vía Alternativa del Complemento , Virus del Sarampión/fisiología , Proteínas Virales de Fusión/fisiología , Animales , Antígenos CD/fisiología , Células CHO , Complemento C3b/fisiología , Factor B del Complemento/fisiología , Cricetinae , Humanos , Proteína Cofactora de Membrana , Glicoproteínas de Membrana/fisiología
19.
J Virol ; 78(15): 7894-903, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15254162

RESUMEN

The trimeric fusion (F) glycoproteins of morbilliviruses are activated by furin cleavage of the precursor F(0) into the F(1) and F(2) subunits. Here we show that an additional membrane-proximal cleavage occurs and modulates F protein function. We initially observed that the ectodomain of approximately one in three measles virus (MV) F proteins is cleaved proximal to the membrane. Processing occurs after cleavage activation of the precursor F(0) into the F(1) and F(2) subunits, producing F(1a) and F(1b) fragments that are incorporated in viral particles. We also detected the F(1b) fragment, including the transmembrane domain and cytoplasmic tail, in cells expressing the canine distemper virus (CDV) or mumps virus F protein. Six membrane-proximal amino acids are necessary for efficient CDV F(1a/b) cleavage. These six amino acids can be exchanged with the corresponding MV F protein residues of different sequence without compromising function. Thus, structural elements of different sequence are functionally exchangeable. Finally, we showed that the alteration of a block of membrane-proximal amino acids results in diminished fusion activity in the context of a recombinant CDV. We envisage that selective loss of the membrane anchor in the external subunits of circularly arranged F protein trimers may disengage them from pulling the membrane centrifugally, thereby facilitating fusion pore formation.


Asunto(s)
Virus del Moquillo Canino/química , Virus del Sarampión/química , Fusión de Membrana , Proteínas Virales de Fusión/química , Secuencia de Aminoácidos , Animales , Chlorocebus aethiops , Virus del Moquillo Canino/fisiología , Retículo Endoplásmico/metabolismo , Virus del Sarampión/fisiología , Datos de Secuencia Molecular , Fragmentos de Péptidos/análisis , Conformación Proteica , Relación Estructura-Actividad , Transfección , Células Vero , Proteínas Virales de Fusión/análisis , Proteínas Virales de Fusión/fisiología
20.
J Virol ; 76(15): 7460-7, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12097558

RESUMEN

The efficiency with which different measles virus (MV) strains enter cells through the immune cell-specific protein SLAM (CD150) or other receptors, including the ubiquitous protein CD46, may influence their pathogenicity. We compared the cell entry efficiency of recombinant MV differing only in their attachment protein hemagglutinin (H). We constructed these viruses with an additional gene expressing an autofluorescent reporter protein to allow direct detection of every infected cell. A virus with a wild-type H protein entered cells through SLAM two to three times more efficiently than a virus with the H protein of the attenuated strain Edmonston, whereas cell entry efficiency through CD46 was lower. However, these subtle differences were amplified at the cell fusion stage because the wild-type H protein failed to fuse CD46-expressing cells. We also proved formally that a mutation in H protein residue 481 (asparagine to tyrosine) results in improved CD46-specific entry. To define the selective pressure exerted on that codon, we monitored its evolution in different H protein backgrounds and found that several passages in CD46-expressing Vero cells were necessary to shift it in the majority of the MV RNA. To verify the importance of these observations for human infections, we examined MV entry into peripheral blood mononuclear cells and observed that viruses with asparagine 481 H proteins infect these cells more efficiently.


Asunto(s)
Antígenos CD/metabolismo , Glicoproteínas/metabolismo , Inmunoglobulinas/metabolismo , Virus del Sarampión/patogenicidad , Glicoproteínas de Membrana/metabolismo , Receptores Virales/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células CHO , Chlorocebus aethiops , Cricetinae , Hemaglutininas Virales/química , Hemaglutininas Virales/genética , Hemaglutininas Virales/metabolismo , Humanos , Leucocitos Mononucleares/virología , Sarampión/virología , Virus del Sarampión/genética , Virus del Sarampión/metabolismo , Proteína Cofactora de Membrana , Fusión de Membrana , Datos de Secuencia Molecular , Receptores de Superficie Celular , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Células Vero
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda