Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(5)2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38473739

RESUMO

A healthy vaginal microbiota hosts Lactobacillus as the most predominant genus. Lactobacilli play a role in human health through the production of diverse antimicrobial substances that can act against human pathogens or modulate the immune system. Previous reports highlighted the ability of vaginal lactobacilli to counteract viruses causing STIs, e.g., HIV-1 and HSV-2. In this report, we analyze the activity of supernatants of vaginal lactobacilli against HSV-1 infection, which is becoming increasingly relevant as a STI. We show that the supernatants of two vaginal Lactobacillus species (i.e., L. crispatus and L. gasseri) were active at neutralizing HSV-1 infection in two different cell lines of human and simian origin. Specifically, we demonstrate that L. crispatus strains are the most effective in antiviral activity, as evidenced by the comparison with a vaginal pathogen taken as reference. The effect was specific and not attributable to the generic toxicity of the supernatants to the cells. Our results pave the way for the development of probiotics to limit the impact of HSV-1 infection on women's health.


Assuntos
Herpes Simples , Herpesvirus Humano 1 , Feminino , Humanos , Lactobacillus , Vagina , Técnicas de Cultura de Células
2.
Viruses ; 13(9)2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34578259

RESUMO

Glioblastoma is a lethal primary brain tumor lacking effective therapy. The secluded onset site, combined with the infiltrative properties of this tumor, require novel targeted therapies. In this scenario, the use of oncolytic viruses retargeted to glioblastoma cells and able to spread across the tumor cells represent an intriguing treatment strategy. Here, we tested the specificity, safety and efficacy of R-613, the first oncolytic HSV fully retargeted to EGFRvIII, a variant of the epidermal growth factor receptor carrying a mutation typically found in glioblastoma. An early treatment with R-613 on orthotopically transplanted EGFRvIII-expressing human glioblastoma significantly increased the median survival time of mice. In this setting, the growth of human glioblastoma xenotransplants was monitored by a secreted luciferase reporter and showed that R-613 is able to substantially delay the development of the tumor masses. When administered as late treatment to a well-established glioblastomas, R-613 appeared to be less effective. Notably the uninfected tumor cells derived from the explanted tumor masses were still susceptible to R-613 infection ex vivo, thus suggesting that multiple treatments could enhance R-613 therapeutic efficacy, making R-613 a promising oncolytic HSV candidate for glioblastoma treatment.


Assuntos
Receptores ErbB/genética , Glioblastoma/terapia , Herpesvirus Humano 1/fisiologia , Terapia Viral Oncolítica/métodos , Terapia Viral Oncolítica/normas , Vírus Oncolíticos/fisiologia , Transplante Heterólogo , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Vetores Genéticos , Humanos , Camundongos , Camundongos SCID , Mutação , Células Vero , Replicação Viral
3.
Int J Mol Sci ; 21(21)2020 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-33167582

RESUMO

Oncolytic viruses are smart therapeutics against cancer due to their potential to replicate and produce the needed therapeutic dose in the tumor, and to their ability to self-exhaust upon tumor clearance. Oncolytic virotherapy strategies based on the herpes simplex virus are reaching their thirties, and a wide variety of approaches has been envisioned and tested in many different models, and on a range of tumor targets. This huge effort has culminated in the primacy of an oncolytic HSV (oHSV) being the first oncolytic virus to be approved by the FDA and EMA for clinical use, for the treatment of advanced melanoma. The path has just been opened; many more cancer types with poor prognosis await effective and innovative therapies, and oHSVs could provide a promising solution, especially as combination therapies and immunovirotherapies. In this review, we analyze the most recent advances in this field, and try to envision the future ahead of oHSVs.


Assuntos
Terapia Viral Oncolítica/métodos , Simplexvirus/metabolismo , Terapia Combinada/métodos , Terapia Combinada/tendências , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Humanos , Terapia Viral Oncolítica/tendências , Vírus Oncolíticos/genética , Vírus Oncolíticos/metabolismo , Simplexvirus/genética
4.
Acta Biomed ; 91(4): e2020144, 2020 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-33525202

RESUMO

BACKGROUND AND AIM: Testing represents one of the main pillars of public health response to SARS-CoV-2/COVID-19 pandemic. This paper shows how accuracy and utility of testing programs depend not just on the type of tests, but on the context as well. METHODS: We describe the testing methods that have been developed and the possible testing strategies; then, we focus on two possible methods of population-wide testing, i.e., pooled testing and testing with rapid antigen tests. We show the accuracy of split-pooling method and how, in different pre-test probability scenarios, the positive and negative predictive values vary using rapid antigen tests. RESULTS: Split-pooling, followed by retesting of negative results, shows a higher sensitivity than individual testing and requires fewer tests. In case of low pre-test probability, a negative result with antigen test could allow to rule out the infection, while, in case of a positive result, a confirmatory molecular test would be necessary. CONCLUSIONS: Test performance alone is not enough to properly choose which test to use; goals and context of the testing program are essential. We advocate the use of pooled strategies when planning population-wide screening, and the weekly use of rapid tests for close periodic monitoring in low-prevalence populations.


Assuntos
Teste para COVID-19 , COVID-19/diagnóstico , Humanos , Valor Preditivo dos Testes , Curva ROC , Reprodutibilidade dos Testes
5.
Oncogene ; 38(23): 4467-4479, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30755732

RESUMO

Oncolytic herpes simplex viruses are proving to be effective in clinical trials against a number of cancers. Here, R-115, an oncolytic herpes simplex virus retargeted to human erbB-2, fully virulent in its target cells, and armed with murine interleukin-12 was evaluated in a murine model of glioblastoma. We show that a single R-115 injection in established tumors resulted, in about 30% of animals, in the complete eradication of the tumor, otherwise invariably lethal. The treatment also induced a significant improvement in the overall median survival time of mice and a resistance to recurrence from the same neoplasia. Such a high degree of protection was unprecedented; it was not observed before following treatments with the commonly used, mutated/attenuated oncolytic viruses. This is the first study providing the evidence of benefits offered by a fully virulent, retargeted, and armed herpes simplex virus in the treatment of glioblastoma and paves the way for clinical translation.


Assuntos
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Imunoterapia/métodos , Terapia Viral Oncolítica/métodos , Simplexvirus/genética , Animais , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Glioblastoma/metabolismo , Humanos , Subunidade p35 da Interleucina-12/metabolismo , Estimativa de Kaplan-Meier , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Recidiva Local de Neoplasia , Vírus Oncolíticos , Proteínas Proto-Oncogênicas c-sis/metabolismo , Receptor ErbB-2/metabolismo , Resultado do Tratamento
6.
Viruses ; 10(7)2018 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-29966356

RESUMO

Previously, we engineered oncolytic herpes simplex viruses (o-HSVs) retargeted to the HER2 (epidermal growth factor receptor 2) tumor cell specific receptor by the insertion of a single chain antibody (scFv) to HER2 in gD, gH, or gB. Here, the insertion of scFvs to three additional cancer targets­EGFR (epidermal growth factor receptor), EGFRvIII, and PSMA (prostate specific membrane antigen)­in gD Δ6­38 enabled the generation of specifically retargeted o-HSVs. Viable recombinants resulted from the insertion of an scFv in place of aa 6­38, but not in place of aa 61­218. Hence, only the gD N-terminus accepted all tested scFv inserts. Additionally, the insertion of mIL12 in the US1-US2 intergenic region of the HER2- or EGFRvIII-retargeted o-HSVs, and the further insertion of Gaussia Luciferase, gave rise to viable recombinants capable of secreting the cytokine and the reporter. Lastly, we engineered two known mutations in gB; they increased the ability of an HER2-retargeted recombinant to spread among murine cells. Altogether, current data show that the o-HSV carrying the aa 6­38 deletion in gD serves as a platform for the specific retargeting of o-HSV tropism to a number of human cancer targets, and the retargeted o-HSVs serve as simultaneous vectors for two molecules.


Assuntos
Expressão Gênica , Genes Reporter , Vetores Genéticos/genética , Vírus Oncolíticos/genética , Simplexvirus/genética , Animais , Antígenos de Superfície/genética , Antígenos de Superfície/metabolismo , Linhagem Celular Tumoral , Chlorocebus aethiops , Receptores ErbB/genética , Receptores ErbB/metabolismo , Ordem dos Genes , Marcação de Genes , Engenharia Genética , Glutamato Carboxipeptidase II/genética , Glutamato Carboxipeptidase II/metabolismo , Humanos , Interleucina-12/genética , Interleucina-12/metabolismo , Camundongos , Mutação , Transgenes , Tropismo Viral , Replicação Viral
7.
Viruses ; 8(3): 63, 2016 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26927159

RESUMO

Most of the oncolytic herpes simplex viruses (HSVs) exhibit a high safety profile achieved through attenuation. They carry defects in virulence proteins that antagonize host cell response to the virus, including innate response, apoptosis, authophagy, and depend on tumor cell proliferation. They grow robustly in cancer cells, provided that these are deficient in host cell responses, which is often the case. To overcome the attenuation limits, a strategy is to render the virus highly cancer-specific, e.g., by retargeting their tropism to cancer-specific receptors, and detargeting from natural receptors. The target we selected is HER-2, overexpressed in breast, ovarian and other cancers. Entry of wt-HSV requires the essential glycoproteins gD, gH/gL and gB. Here, we reviewed that oncolytic HSV retargeting was achieved through modifications in gD: the addition of a single-chain antibody (scFv) to HER-2 coupled with appropriate deletions to remove part of the natural receptors' binding sites. Recently, we showed that also gH/gL can be a retargeting tool. The insertion of an scFv to HER-2 at the gH N-terminus, coupled with deletions in gD, led to a recombinant capable to use HER-2 as the sole receptor. The retargeted oncolytic HSVs can be administered systemically by means of carrier cells-forcedly-infected mesenchymal stem cells. Altogether, the retargeted oncolytic HSVs are highly cancer-specific and their replication is not dependent on intrinsic defects of the tumor cells. They might be further modified to express immunomodulatory molecules.


Assuntos
Vírus Oncolíticos/fisiologia , Receptor ErbB-2/metabolismo , Receptores Virais/metabolismo , Simplexvirus/fisiologia , Proteínas Estruturais Virais/metabolismo , Tropismo Viral , Vírus Oncolíticos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Simplexvirus/genética , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/metabolismo , Proteínas Estruturais Virais/genética , Internalização do Vírus
8.
Curr Opin Virol ; 2(1): 28-36, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22440963

RESUMO

Herpes simplex virus (HSV) entry into the cell involves the fusion of the virion envelope with a cellular membrane and delivery of capsid and tegument proteins to the cytoplasm. Our understanding of this phenomenon has greatly increased in recent years. On the virus side, the multipartite nature of the entry-fusion machinery (made of the glycoproteins gD, the heterodimer gH/gL and gB) entails a mechanism of gD activation promoted by the gD encounter with one of its receptor; and cross-talk among the entry-fusion glycoproteins, which culminates in gB activation and fusion execution. On the cell side, machineries and signalling activities are put in place. The number of known receptors and sentinels is increasing. The cell routes the virus through alternative entry pathways by means of routing factors, exemplified by αVß3-integrin and paired immunoglobulin-like type 2 receptor alpha. Of the signalling events, a key one is the immediate host response to incoming virions. Unexpectedly, this is in part triggered by the same virion components and some cellular factors that also promote virus entry. Hence, a link is emerging between two phenomena so far considered as distinct.


Assuntos
Herpes Simples/metabolismo , Simplexvirus/fisiologia , Internalização do Vírus , Animais , Herpes Simples/genética , Herpes Simples/virologia , Humanos , Simplexvirus/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
9.
J Virol ; 83(20): 10752-60, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19656900

RESUMO

The gD, gB, and gH/gL glycoprotein quartet constitutes the basic apparatus for herpes simplex virus (HSV) entry into the cell and fusion. gD serves as a receptor binding glycoprotein and trigger of fusion. The conserved gB and gH/gL execute fusion. Central to understanding HSV entry/fusion has become the dissection of how the four glycoproteins engage in cross talk. While the independent interactions of gD with gB and gD with gH/gL have been documented, less is known of the interaction of gB with gH/gL. So far, this interaction has been detected only in the presence of gD by means of a split green fluorescent protein complementation assay. Here, we show that gB interacts with gH/gL in the absence of gD. The gB-gH/gL complex was best detected with a form of gB in which the endocytosis and phosphorylation motif have been deleted; this form of gB persists in the membranes of the exocytic pathway and is not endocytosed. The gB-gH/gL interaction was detected both in whole transfected cells by means of a split yellow fluorescent protein complementation assay and, biochemically, by a pull-down assay. Results with a panel of chimeric forms of gB, in which portions of the glycoprotein bracketed by consecutive cysteines were replaced with the corresponding portions from human herpesvirus 8 gB, favor the view that gB carries multiple sites for interaction with gH/gL, and one of these sites is located in the pleckstrin-like domain 1 carrying the bipartite fusion loop.


Assuntos
Simplexvirus , Proteínas do Envelope Viral/metabolismo , Animais , Células COS , Linhagem Celular , Chlorocebus aethiops , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Fusão de Membrana , Simplexvirus/genética , Simplexvirus/metabolismo , Simplexvirus/patogenicidade , Simplexvirus/fisiologia , Proteínas do Envelope Viral/genética , Internalização do Vírus
10.
J Virol ; 81(20): 11532-7, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17670828

RESUMO

The interactions between herpes simplex virus gD and its nectin1 receptor or between gD, gB, and gH were analyzed by complementation of the N and C portions of split enhanced green fluorescent protein (EGFP) fused to the glycoproteins. The gD(N)-Nect(C) complex was readily detected; the gD(N)-gC(C) complex was undetectable, highlighting the specificity of the assay. Split EGFP complementation was detected between proteins designated gD(N)+gH(C), gD(N)+gB(C), and gH(N)+gB(C)+wtgD (gB was deleted of endocytosis motifs), both in cells transfected with two-tree glycoproteins and in syncytia. The in situ assay provides evidence that gD interacts with gH and gB independently of each other and supports a model whereby gH and gB in complex exert their activities to gD.


Assuntos
Simplexvirus/química , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Glicoproteínas/metabolismo , Proteínas de Fluorescência Verde , Ligação Proteica , Proteínas Recombinantes de Fusão , Simplexvirus/patogenicidade , Transdução Genética
11.
Rev Med Virol ; 17(5): 313-26, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17573668

RESUMO

The multipartite entry-fusion system of herpes simplex virus is made of a quartet of glycoproteins-gD, gB, gH.gL-and three alternative gD receptors, herpesvirus entry mediator (HVEM), nectin1 and modified sites on heparan sulphate. This multipartite system recapitulates the basic steps of virus-cell fusion, i.e. receptor recognition, triggering of fusion and fusion execution. Specifically, in addition to serving as the receptor-binding glycoprotein, gD triggers fusion through a specialised domain, named pro-fusion domain (PFD), located C-terminally in the ectodomain. In the unliganded gD the C-terminal region folds around the N-terminal region, such that gD adopts a closed autoinhibited conformation. In HVEM- and nectin1-bound gD the C-terminal region is displaced (opened conformation). gD is the tool for modification of HSV tropism, through insertion of ligands to heterologous tumour-specific receptors. It is discussed whether gD responds to the interaction with the natural and the heterologous receptors by adopting similar conformations, and whether the closed-to-open switch in conformation is a generalised mechanism of activation. A peculiar recombinant highlighted that the central Ig-folded core of gD may not encode executable functions for entry and that the 219-314 aa segment may be sufficient to trigger fusion. With respect to fusion execution, gB appears to be a prospective fusogen based on its coiled-coil trimeric structure, similar to that of another fusion glycoprotein. On the other hand, gH exhibits molecular elements typical of class 1 fusion glycoproteins, in particular heptad repeats and strong tendency to interact with lipids. Whether fusion execution is carried out by gB or gH.gL, or both glycoproteins in complex or sequentially remains to be determined.


Assuntos
Simplexvirus/fisiologia , Internalização do Vírus , Glicoproteínas/química , Glicoproteínas/fisiologia , Humanos , Modelos Moleculares , Receptores Virais/fisiologia , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/fisiologia
12.
J Virol ; 78(15): 8015-25, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15254173

RESUMO

Syncytium formation in cells that express herpes simplex virus glycoprotein B (gB), gD, gH, and gL is blocked by gK (E. Avitabile, G. Lombardi, and G. Campadelli-Fiume, J. Virol. 77:6836-6844, 2003). Here, we report the results of two series of experiments. First, UL20 protein (UL20p) expression weakly inhibited cell-cell fusion. Coexpression of UL20p and gK drastically reduced fusion in a cell-line-dependent manner, with the highest inhibition in BHK cells. Singly expressed UL20p and gK localized at the endoplasmic reticulum and nuclear membranes. When they were coexpressed, both proteins relocalized to the Golgi apparatus. Remarkably, in cells that coexpressed UL20p and gK, the antifusion activity correlated with a downmodulation of gD, gB, gH, and gL cell surface expression. Second, gB(Delta867) has a partial deletion in the cytoplasmic tail that removed endocytosis motifs. Whereas wild-type (wt) gB was internalized in vesicles lined with the endosomal marker Rab5, gB(delta867) was not internalized, exhibited enhanced cell surface expression, and was more efficient in mediating cell-cell fusion than wt gB. The antifusion activity of UL20p and gK was also exerted when gB(delta867) replaced wt gB in the cell fusion assay. These studies show that the gB C tail carries a functional endocytosis motif(s) and that the removal of the motif correlated with increased gB surface expression and increased fusion activity. We conclude that cell-cell fusion in wt-virus-infected cells is negatively controlled by at least two mechanisms. The novel mechanism described here involves the concerted action of UL20p and gK and correlates with a moderate but consistent reduction in the cell surface expression of the fusion glycoproteins. This mechanism is independent of the one exerted through endocytosis-mediated downmodulation of gB from the plasma membrane.


Assuntos
Endocitose , Fusão de Membrana , Proteínas do Envelope Viral/fisiologia , Proteínas Virais/fisiologia , Animais , Linhagem Celular , Regulação para Baixo
13.
J Biol Chem ; 278(38): 36059-67, 2003 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-12844494

RESUMO

Signals involved in protection against apoptosis by herpes simplex virus 1 (HSV-1) were investigated. Using U937 monocytoid cells as an experimental model, we have demonstrated that HSV-1 rendered these cells resistant to Fas-induced apoptosis promptly after infection. UV-inactivated virus as well as the envelope glycoprotein D (gD) of HSV-1, by itself, exerted a protective effect on Fas-induced apoptosis. NF-kappaB was activated by gD, and protection against Fas-mediated apoptosis by gD was abolished in cells stably transfected with a dominant negative mutant I-kappaBalpha, indicating that NF-kappaB activation plays a role in the antiapoptotic activity of gD in our experimental model. Moreover, NF-kappaB-dependent protection against Fas-mediated apoptosis was associated with decreased levels of caspase-8 activity and with the up-regulation of intracellular antiapoptotic proteins.


Assuntos
Apoptose , NF-kappa B/fisiologia , Proteínas do Envelope Viral/fisiologia , Receptor fas/metabolismo , Animais , Western Blotting , Caspase 8 , Caspase 9 , Caspases/metabolismo , Técnicas de Cocultura , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Genes Dominantes , Humanos , Proteínas I-kappa B/metabolismo , Marcação In Situ das Extremidades Cortadas , Cinética , Camundongos , Mutação , Inibidor de NF-kappaB alfa , NF-kappa B/metabolismo , Fatores de Tempo , Transfecção , Células U937 , Raios Ultravioleta , Regulação para Cima , Proteínas do Envelope Viral/metabolismo
14.
J Virol ; 77(12): 6836-44, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12768003

RESUMO

A Myc epitope was inserted at residue 283 of herpes simplex virus type 1 (HSV-1) glycoprotein K (gK), a position previously shown not to interfere with gK activity. The Myc-tagged gK localized predominantly to the endoplasmic reticulum, both in uninfected and in HSV-infected cells. gK, coexpressed with the four HSV fusogenic glycoproteins, gD, gB, gH, and gL, inhibited cell-cell fusion. The effect was partially dose dependent and was observed both in baby hamster kidney (BHK) and in Vero cells, indicating that the antifusion activity of gK may be cell line independent. The antifusion activity of gK did not require viral proteins other than the four fusogenic glycoproteins. A syncytial (syn) allele of gK (syn-gK) carrying the A40V substitution present in HSV-1(MP) did not block fusion to the extent seen with the wild-type (wt) gK, indicating that the syn mutation ablated, at least in part, the antifusogenic activity of wt gK. We conclude that gK is part of the mechanism whereby HSV negatively regulates its own fusion activity. Its effect accounts for the notion that cells infected with wt HSV do not fuse with adjacent, uninfected cells into multinucleated giant cells or syncytia. gK may also function to preclude fusion between virion envelope and the virion-encasing vesicles during virus transport to the extracellular compartment, thus preventing nucleocapsid de-envelopment in the cytoplasm.


Assuntos
Alelos , Células Gigantes , Herpesvirus Humano 1/fisiologia , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/farmacologia , Animais , Fusão Celular , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Células Gigantes/fisiologia , Herpesvirus Humano 1/genética , Humanos , Fusão de Membrana , Células Vero , Proteínas do Envelope Viral/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo
15.
J Virol ; 77(6): 3759-67, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12610150

RESUMO

Glycoprotein D (gD) interacts with two alternative protein receptors, nectin1 and HveA, to mediate herpes simplex virus (HSV) entry into cells. Fusion of the envelope with the plasma membrane requires, in addition to gD, glycoproteins gB, gH, and gL. Coexpression of the four glycoproteins (gD, gB, gH, and gL) promotes cell-cell fusion. gD delivered in trans is also capable of blocking the apoptosis induced by gD deletion viruses grown either in noncomplementing cells (gD(-/-)) or in complementing cells (gD(-/+)). While ectopic expression of cation-independent mannose-6 phosphate receptor blocks apoptosis induced by both stocks, other requirements differ. Thus, apoptosis induced by gD(-/-) virus is blocked by full-length gD (or two gD fragments reconstituting a full-length molecule), whereas ectopic expression of the gD ectodomain is sufficient to block apoptosis induced by gD(-/+) virus. In this report we took advantage of a set of gD insertion-deletion mutants to map the domains of gD required to block apoptosis by gD(-/-) and gD(-/+) viruses and those involved in cell-cell fusion. The mutations that resulted in failure to block apoptosis were the same for gD(-/-) and gD(-/+) viruses and were located in three sites, one within the immunoglobulin-type core region (residues 125, 126, and 151), one in the upstream connector region (residues 34 and 43), and one in the C-terminal portion of the ectodomain (residue 277). A mutant that carried amino acid substitutions at the three glycosylation sites failed to block apoptosis but behaved like wild-type gD in all other assays. The mutations that inhibited polykaryocyte formation were located in the upstream connector region (residues 34 and 43), at the alpha1 helix (residue 77), in the immunoglobulin core and downstream regions (residue 151 and 187), and at the alpha3 helix (residues 243 and 246). Binding of soluble nectin1-Fc to cells expressing the mutant gDs was generally affected by the same mutations that affected fusion, with one notable exception (Delta277-310), which affected fusion without hampering nectin1 binding. This deletion likely identifies a region of gD involved in fusion activity at a post-nectin1-binding step. We conclude that whereas mutations that affected all functions (e.g., upstream connector region and residue 151) may be detrimental to overall gD structure, the mutations that affect specific activities identify domains of gD involved in the interactions with entry receptors and fusogenic glycoproteins and with cellular proteins required to block apoptosis. The evidence that glycosylation of gD is required for blocking apoptosis supports the conclusion that the interacting protein is the mannose-6 phosphate receptor.


Assuntos
Apoptose , Moléculas de Adesão Celular/metabolismo , Fusão Celular , Herpesvirus Humano 1/patogenicidade , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Animais , Linhagem Celular , Cricetinae , Humanos , Mutação , Nectinas , Receptores Virais/metabolismo , Spodoptera , Proteínas do Envelope Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...