Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Viral Immunol ; 18(2): 317-26, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16035943

RESUMO

Live attenuated RNA viruses make highly efficient vaccines. Among them is the live attenuated measles virus (MV) vaccine that has been given to a very large number of children and has been shown to be highly efficacious and safe. MV vaccine induces a life-long immunity after a single injection or two low-dose injections. It is easily produced on a large scale in most countries and can be distributed at low cost. Reversion to pathogenicity has never been observed with this vaccine. For all of these characteristics, developing of MV vaccine vector as a multivalent vaccine to immunize children against both measles and other infectious agents such as human immunodeficiency virus (HIV), flaviviruses, or malaria might be very promising for worldwide use. As MV vaccine is inexpensive to produce, the generation of recombinant vaccines may remain affordable and attractive for the developing word. In this article, we describe the development of MV vector and present some recent data showing the capacity of recombinant MV vaccine to express various proteins from HIV and West Nile virus. In addition, the ability of recombinant MV to induce specific immune responses against these different pathogens are presented and discussed.


Assuntos
Vetores Genéticos , HIV-1 , Vacina contra Sarampo , Vacinas Atenuadas , Proteínas do Envelope Viral/metabolismo , Vírus do Nilo Ocidental , Criança , Vetores Genéticos/administração & dosagem , Vetores Genéticos/imunologia , Infecções por HIV/imunologia , Infecções por HIV/prevenção & controle , HIV-1/genética , HIV-1/imunologia , Humanos , Sarampo/prevenção & controle , Vacina contra Sarampo/administração & dosagem , Vacina contra Sarampo/genética , Vacina contra Sarampo/imunologia , Vírus do Sarampo/genética , Vírus do Sarampo/imunologia , Vírus do Sarampo/metabolismo , Recombinação Genética , Vacinação , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Febre do Nilo Ocidental/prevenção & controle , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/imunologia
2.
Hum Vaccin Immunother ; 11(1): 21-6, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25483511

RESUMO

Measles was an inevitable infection during the human development with substantial degree of morbidity and mortality. The severity of measles virus (MV) infection was largely contained by the development of a live attenuated vaccine that was introduced into the vaccination programs. However, all efforts to eradicate the disease failed and continued to annually result in significant deaths. The development of molecular biology techniques allowed the rescue of MV from cDNA that enabled important insights into a variety of aspects of the biology of the virus and its pathogenesis. Subsequently these technologies facilitated the development of novel vaccine candidates that induce immunity against measles and other pathogens. Based on the promising prospective, the use of MV as a recombinant vaccine and a therapeutic vector is addressed.


Assuntos
Portadores de Fármacos , Vetores Genéticos , Vacina contra Sarampo/imunologia , Vírus do Sarampo/imunologia , Vírus do Sarampo/fisiologia , Humanos , Sarampo/epidemiologia , Sarampo/prevenção & controle , Vacina contra Sarampo/administração & dosagem , Vacina contra Sarampo/genética , Vírus do Sarampo/genética , Terapia Viral Oncolítica/métodos , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
3.
Hum Vaccin Immunother ; 11(1): 83-90, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25483519

RESUMO

Today, immune compromised interferon-α-receptor deficient mice expressing hCD46 (IFNARCD46tg) are usually used for measles virus (MV) based vaccine characterization. However, for the development of MV-based recombinant vaccine candidates (rMV), an immune competent mouse model is desirable in order to induce and evaluate meaningful immune response. In this study, humoral and cellular immune response induced by rMV in immune competent mice expressing human MV receptor CD46 (hCD46tg) were compared with those induced in wild-type black/6, and IFNARCD46tg mice.   All three strains developed humoral and cellular response against MV, whereas only hCD46tg and IFNARCD46tg mice developed a humoral response against the transgene. Differences were observed in the magnitude of the response, where the IFNARCD46tg mice displayed the strongest immune responses, followed by the hCD46tg mice and the black/6 mice. Interestingly, hCD46tg and wt black/6 mice showed a predominant CD4(+) T-cell response against MV-N, whereas IFNARCD46tg mice developed both, CD4(+) and CD8(+) T-cell response against MV-N. Analysis of the cytokine profile of MV-N specific CD4(+) T-cells and transgene (SIVgag) specific CD8(+) T-cells revealed qualitative differences of the T-cell responses; noticeably a significant reduction of the frequency of CD4(+)IL-2(+) expressing cells in IFNARCD46tg mice as compared with hCD46tg or wt black/6 mice. We show in this study significant quantitative and qualitative differences in immune responses between immune competent and immune-compromised mice. Our results therefore highlight the importance of the animal model and support the use of hCD46tg mice as mouse model for the characterization of the immunological profile induced by recombinant measles virus vaccine candidates.


Assuntos
Vacina contra Sarampo/imunologia , Vírus do Sarampo/imunologia , Modelos Animais , Animais , Anticorpos Antivirais/sangue , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Citocinas/metabolismo , Feminino , Expressão Gênica , Masculino , Sarampo/imunologia , Sarampo/prevenção & controle , Vacina contra Sarampo/administração & dosagem , Vacina contra Sarampo/genética , Vírus do Sarampo/genética , Proteína Cofatora de Membrana/genética , Camundongos , Camundongos Transgênicos , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
4.
Hum Vaccin Immunother ; 9(3): 457-61, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23249651

RESUMO

The exceptional discoveries of antigen/gene delivery systems have allowed the development of novel prophylactic and therapeutic vaccine candidates. The vaccine candidates employ various antigen-delivery systems, particularly recombinant viral vectors. Recombinant viral vectors are experimental vaccines similar to DNA vaccines, but they use attenuated viruses or bacterium as a carrier "vector" to introduce microbial DNA to cells of the body. They closely mimic a natural infection and therefore can efficiently stimulate the immune system. Although such recombinant vectors may face extensive preclinical testing and will possibly have to meet stringent regulatory requirements, some of these vectors (e.g. measles virus vectors) may benefit from the profound industrial and clinical experience of the parent vaccine. Most notably, novel vaccines based on live attenuated viruses combine the induction of broad, strong and persistent immune responses with acceptable safety profiles. We assess certain technologies in light of their use against human immunodeficiency virus (HIV).


Assuntos
Portadores de Fármacos , Descoberta de Drogas/métodos , Vetores Genéticos , Vírus do Sarampo/genética , Vacinas Virais/imunologia , Vacinas Virais/isolamento & purificação , Descoberta de Drogas/tendências , Humanos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/isolamento & purificação , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Vacinas de DNA/isolamento & purificação , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/isolamento & purificação , Vacinas de Partículas Semelhantes a Vírus/genética , Vacinas de Partículas Semelhantes a Vírus/imunologia , Vacinas de Partículas Semelhantes a Vírus/isolamento & purificação , Vacinas Virais/genética
5.
Hum Vaccin Immunother ; 9(3): 599-606, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23324399

RESUMO

Measles virus (MV) vectors are promising candidates for designing new recombinant vaccines since the parental live vaccines have a well-known safety and efficacy record. Like all viral vectors, the MV vector efficacy in inducing a protecting immune answer could be affected by the pre-existing immunity among the human population. In order to determine the optimal immunization route and regimen, we mimicked a MV pre-immunity by passively administrating MV neutralizing antibodies (MV-nAb) prior intramuscular (i.m.) and/or intranasal (i.n.) immunization with recombinant MV expressing the SIV-gag antigen (rMV-SIVgag). Our results revealed that 500 mIU of MV-nAb allowed the induction of a humoral and cellular immune response against the vector and the transgene, while higher titers of the MV-nAb were significantly inhibitory. In a prime-boost regimen, in the presence of MV-nAb, the intranasal-intramuscular (i.n.-i.m.) or intramuscular-intramuscular (i.m.-i.m.) routes induced higher humoral immune responses against the vector and the transgene (SIV-gag). In naive animals, cellular immune response was significantly higher by i.m. immunization; however, MV pre-immunity did not seem to affect the cellular immune response after an i.n. immunization.   In summary, we show that a pre-existing immunity of up to 500 mIU anti-MV neutralizing antibodies had little effect on the replication of rMV and did not inhibit the induction of significant humoral and cellular immune responses in immune-competent mice.


Assuntos
Anticorpos Antivirais/sangue , Portadores de Fármacos , Vetores Genéticos/imunologia , Imunização/métodos , Vírus do Sarampo/imunologia , Sarampo/imunologia , Vacinas Virais/imunologia , Administração Intranasal , Animais , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Antivirais/administração & dosagem , Humanos , Imunidade Celular , Injeções Intramusculares , Vírus do Sarampo/genética , Camundongos , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
6.
Hum Vaccin Immunother ; 9(3): 607-13, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23324616

RESUMO

The measles virus vaccine (MVbv) is a clinically certified and well-tolerated vaccine strain that has been given both parenterally and mucosally. It has been extensively used in children and has proven to be safe and effective in eliciting protective immunity. This specific strain was therefore chosen to generate a measles viral vector. The genome of the commercial MVbv vaccine strain was isolated, sequenced and a plasmid, p(+)MVb, enabling transcription of the viral antigenome and rescue of MVb, was constructed. Phylogenic and phenotypic analysis revealed that MVbv and the rescued MVb constitute another evolutionary branch within the hitherto classified measles vaccines. Plasmid p(+)MVb was modified by insertion of artificial MV-type transcription units (ATUs) for the generation of recombinant viruses (rMVb) expressing additional proteins. Replication characteristics and immunogenicity of rMVb vectors were similar to the parental MVbv and to other vaccine strains. The expression of the additional proteins was stable over 10 serial virus transfers, which corresponds to an amplification greater than 10 ( 20) . The excellent safety record and its efficient application as aerosol may add to the usefulness of the derived vectors.


Assuntos
Vetores Genéticos , Vírus do Sarampo/genética , Vacinas Virais/imunologia , Animais , Chlorocebus aethiops , Análise por Conglomerados , Expressão Gênica , Instabilidade Genômica , Dados de Sequência Molecular , Filogenia , Plasmídeos , Análise de Sequência de DNA , Homologia de Sequência , Células Vero , Vacinas Virais/genética , Replicação Viral
7.
Vaccine ; 27(25-26): 3385-90, 2009 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-19200837

RESUMO

Cervical cancer is mainly associated with HPV genotype 16 infection. Recombinant measles virus (rMV) expressing HPV genotype 16 L1 capsid protein was generated by construction of an antigenomic plasmid, followed by rescue using the human "helper" cell line 293-3-46. In cell cultures the recombinant MV-L1 virus replicated practically as efficiently as the standard attenuated MV established as commercial vaccine, devoid of the transgene. The high genetic stability of MVb2-L1 was confirmed by 10 serial viral transfers in cell culture. In transgenic mice expressing the MV receptor CD46 the recombinant induced strong humoral immune responses against both MV and HPV; the antibodies against L1 exhibited mainly neutralizing capacity. Our data suggest that MV is a promising vehicle for development of inexpensive and efficient vaccines protecting from HPV infection.


Assuntos
Proteínas do Capsídeo/imunologia , Vírus do Sarampo/genética , Proteínas Oncogênicas Virais/imunologia , Vacinas contra Papillomavirus/imunologia , Neoplasias do Colo do Útero/prevenção & controle , Vacinas Sintéticas/imunologia , Animais , Anticorpos Antivirais/sangue , Sequência de Bases , Proteínas do Capsídeo/genética , Chlorocebus aethiops , Ensaio de Imunoadsorção Enzimática , Feminino , Camundongos , Dados de Sequência Molecular , Proteínas Oncogênicas Virais/genética , Proteínas Recombinantes/imunologia , Células Vero , Replicação Viral
8.
Vaccine ; 27(25-26): 3299-305, 2009 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-19200842

RESUMO

Recombinant measles viruses (rMV) based on the live attenuated measles vaccine strain (MVb) expressing antigens of HIV-1 clade B were generated by reverse genetics. Recombinants expressing single or double antigens of HIV-1 (rMV-HIV) were genetically highly stable on human diploid cells. The production process of these viruses was essentially similar to the parental MV strain, yielding comparative end titers. Immunization of tg-mice by different regimens and formulations showed potent humoral and cellular immune responses against MV and HIV antigens. Recombinant MV-HIV expressing Gag protein conferred protective immunity in tg-mice after a high-dose pseudochallenge with recombinant vaccinia virus. In addition, rMV-HIV boosted anti-HIV antibodies, in the presence of pre-existing anti-vector antibodies.


Assuntos
Vacinas contra a AIDS/imunologia , Antígenos HIV/imunologia , HIV-1/imunologia , Vírus do Sarampo/genética , Vacinas Sintéticas/imunologia , Animais , Anticorpos Antivirais/sangue , Antígenos HIV/genética , HIV-1/genética , Humanos , Interferon gama/biossíntese , Vírus do Sarampo/imunologia , Camundongos , Camundongos Transgênicos , Proteínas Recombinantes/biossíntese , Transgenes
9.
Vaccine ; 26(17): 2164-74, 2008 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-18346823

RESUMO

Live attenuated recombinant measles viruses (rMV) expressing a codon-optimised spike glycoprotein (S) or nucleocapsid protein (N) of severe acute respiratory syndrome-associated coronavirus (SARS-CoV) were generated (rMV-S and rMV-N). Both recombinant viruses stably expressed the corresponding SARS-CoV proteins, grew to similar end titres as the parental strain and induced high antibody titres against MV and the vectored SARS-CoV antigens (S and N) in transgenic mice susceptible to measles infection. The antibodies induced by rMV-S had a high neutralising effect on SARS-CoV as well as on MV. Moreover, significant N-specific cellular immune responses were measured by IFN-gamma ELISPOT assays. The pre-existence of anti-MV antibodies induced by the initial immunisation dose did not inhibit boost of anti-S and anti-N antibodies. Immunisations comprising a mixture of rMV-S and rMV-N induced immune responses similar in magnitude to that of vaccine components administered separately. These data support the suitability of MV as a bivalent candidate vaccine vector against MV and emerging viruses such as SARS-CoV.


Assuntos
Vírus do Sarampo/fisiologia , Glicoproteínas de Membrana/imunologia , Síndrome Respiratória Aguda Grave/prevenção & controle , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/administração & dosagem , Animais , Animais Geneticamente Modificados , Vetores Genéticos/química , Vacina contra Sarampo/administração & dosagem , Vacina contra Sarampo/genética , Vacina contra Sarampo/imunologia , Vírus do Sarampo/metabolismo , Glicoproteínas de Membrana/genética , Camundongos , Testes de Neutralização , Proteínas do Nucleocapsídeo/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/imunologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/metabolismo , Síndrome Respiratória Aguda Grave/imunologia , Glicoproteína da Espícula de Coronavírus , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Vacinas Virais/química , Vacinas Virais/genética , Vacinas Virais/imunologia
10.
Expert Rev Vaccines ; 6(2): 255-66, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17408374

RESUMO

The exceptional discoveries of antigen/gene delivery systems have allowed the development of novel prophylactic and therapeutic vaccine candidates. This review highlights various antigen-delivery systems, particularly viral vectors, and assesses the underlying technologies in light of their use against AIDS and malaria. Although such recombinant vectors may face extensive preclinical testing and will possibly have to meet stringent regulatory requirements, some of these vectors may benefit from the profound industrial and clinical experience of the parent vaccine. Most notably, novel vaccines based on live, recombinant vectors may combine the induction of broad, strong and persistent immune responses with acceptable safety profiles.


Assuntos
Antígenos/imunologia , Vírus de DNA/imunologia , Desenho de Fármacos , Vetores Genéticos/imunologia , Vírus de RNA/imunologia , Vacinas de DNA/imunologia , Vacinas contra a AIDS/imunologia , Adenovírus Humanos/imunologia , Alphavirus/imunologia , Animais , Antígenos/genética , Antígenos de Protozoários/imunologia , Vírus de DNA/genética , Antígenos HIV/imunologia , Herpesvirus Humano 1/imunologia , Humanos , Malária/imunologia , Vacinas Antimaláricas/imunologia , Vírus do Sarampo/imunologia , Poliovirus/imunologia , Poxviridae/imunologia , Vírus de RNA/genética , Vacinas de DNA/genética , Vírus da Estomatite Vesicular Indiana/imunologia , Vírus da Febre Amarela/imunologia
11.
Vaccine ; 25(16): 2974-83, 2007 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-17303293

RESUMO

Live attenuated measles virus (MV) vaccines have an impressive record of safety, efficacy and ability to induce life-long immunity against measles infection. Using reverse genetics technology, such negative-strand RNA viruses can now be rescued from cloned DNA. This technology allows the insertion of exogenous genes encoding foreign antigens into the MV genome in such a way that they can be expressed by the MV vaccine strain, without affecting virus structure, propagation and cell targeting. Recombinant viruses rescued from cloned cDNA induce immune responses against both measles virus and the cloned antigens. The tolerability of MV to gene(s) insertion makes it an attractive flexible vector system, especially if broad immune responses are required. The fact that measles replication strictly occurs in the cytoplasm of infected cells without DNA intermediate has important biosafety implications and adds to the attractiveness of MV as a vector. In this article we report the characteristics of reporter gene expression (GFP, LacZ and CAT) and the biochemical, biophysical and immunological properties of recombinant MV expressing heterologous antigens of simian immunogeficiency virus (SIV).


Assuntos
Antígenos Virais/metabolismo , Vacina contra Sarampo/imunologia , Vírus do Sarampo/imunologia , Sarampo/prevenção & controle , Vacinas Atenuadas/administração & dosagem , Animais , Antígenos Virais/genética , Linhagem Celular , Chlorocebus aethiops , Vetores Genéticos , Sarampo/virologia , Vacina contra Sarampo/genética , Vírus do Sarampo/genética , Vírus do Sarampo/crescimento & desenvolvimento , Vírus do Sarampo/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Células Vero
12.
Vaccine ; 25(14): 2567-74, 2007 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-16914237

RESUMO

A workshop on viral vectors for malaria vaccine development, organized by the PATH Malaria Vaccine Initiative, was held in Bethesda, MD on October 20, 2005. Recent advancements in viral-vectored malaria vaccine development and emerging vector technologies were presented and discussed. Classic viral vectors such as poxvirus, adenovirus and alphavirus vectors have been successfully used to deliver malaria antigens. Some of the vaccine candidates have demonstrated their potential in inducing malaria-specific immunity in animal models and human trials. In addition, emerging viral-vector technologies, such as measles virus (MV), vesicular stomatitis virus (VSV) and yellow fever (YF) virus, may also be useful for malaria vaccine development. Studies in animal models suggest that each viral vector is unique in its ability to induce humoral and/or cellular immune responses. Those studies have also revealed that optimization of Plasmodium genes for mammalian expression is an important aspect of vaccine design. Codon-optimization, surface-trafficking, de-glycosylation and removal of toxic domains can lead to improved immunogenicity. Understanding the vector's ability to induce an immune response and the expression of malaria antigens in mammalian cells will be critical in designing the next generation of viral-vectored malaria vaccines.


Assuntos
Vetores Genéticos/genética , Vacinas Antimaláricas/biossíntese , Vacinas Sintéticas/biossíntese , Vírus/genética , Adenoviridae/genética , Alphavirus/genética , Vacinas Antimaláricas/imunologia , Vírus do Sarampo/genética , Poxviridae/genética , Vacinas Sintéticas/imunologia , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Febre Amarela/genética
13.
J Invest Dermatol ; 126(11): 2525-32, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16960554

RESUMO

Measles virus (MV) has shown promise as an oncolytic virus in the treatment of different tumor models for human B-cell lymphoma, multiple myeloma, ovarian cancer, and glioma. We have shown that, in a phase I clinical trial, MV vaccine induces tumor regression in cutaneous T-cell lymphoma (CTCL) patients. Here, we investigated in detail, the effect of recombinant MV (rMV) vaccine strain in CTCL cell cultures, and in vivo in established CTCL xenografts in nude mice. The susceptibility of three CTCL cell lines, originating from patients, to rMV was tested by determination of cell surface expression of MV receptors. All cell lines expressed the receptors CD150 and CD46 and were easily infected by rMV and induced complete cell lysis. The cytoreductive activity was apparent in cells forming aggregates, indicating a cell-to-cell spread of MV and cytolysis owing to virus infection. Intratumoral (i.t.) injection of rMV, expressing enhanced green fluorescent protein induced complete regression of large established human CTCL tumors in nude mice, whereas tumors with control treatment progressed exponentially. Immunohistochemical analysis of tumor biopsies, after i.t. treatment, for MV-NP protein complex demonstrated replication of MV within the tumors. The data demonstrate the potential of MV as a therapeutic agent against CTCL.


Assuntos
Vacinas Anticâncer/uso terapêutico , Linfoma Cutâneo de Células T/terapia , Vacina contra Sarampo/uso terapêutico , Vírus do Sarampo , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos , Animais , Antígenos CD/análise , Linhagem Celular Tumoral , Humanos , Linfoma Cutâneo de Células T/química , Vírus do Sarampo/genética , Vírus do Sarampo/fisiologia , Proteína Cofatora de Membrana/análise , Camundongos , Camundongos Nus , Vírus Oncolíticos/genética , Vírus Oncolíticos/fisiologia , Receptores de Superfície Celular/análise , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Ativação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
14.
J Virol ; 76(11): 5720-8, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11992000

RESUMO

Measles virus (MV) can infect the central nervous system and, in rare cases, causes subacute sclerosing panencephalitis, characterized by a progressive degeneration of neurons. The route of MV transmission in neurons was investigated in cultured rat hippocampal slices by using MV expressing green fluorescent protein. MV infected hippocampal neurons and spread unidirectionally, in a retrograde manner, from CA1 to CA3 pyramidal cells and from there to the dentate gyrus. Spreading of infection depended on cell-to-cell contact and occurred without any detectable release of infectious particles. The role of the viral proteins in the retrograde MV transmission was determined by investigating their sorting in infected pyramidal cells. MV glycoproteins, the fusion protein (F) and hemagglutinin (H), the matrix protein (M), and the phosphoprotein (P), which is part of the viral ribonucleoprotein complex, were all sorted to the dendrites. While M, P, and H proteins remained more intracellular, the F protein localized to prominent, spine-type domains at the surface of infected cells. The detected localization of MV proteins suggests that local microfusion events may be mediated by the F protein at sites of synaptic contacts and is consistent with a mechanism of retrograde transmission of MV infection.


Assuntos
Hipocampo/virologia , Vírus do Sarampo/fisiologia , Neurônios/virologia , Animais , Comunicação Celular , Chlorocebus aethiops , Técnicas de Cultura , Hemaglutininas Virais/metabolismo , Hipocampo/citologia , Humanos , Vírus do Sarampo/crescimento & desenvolvimento , Vírus do Sarampo/metabolismo , Fosfoproteínas/metabolismo , Ratos , Células Vero , Proteínas Virais de Fusão/metabolismo , Proteínas da Matriz Viral/metabolismo , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa