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
2.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27795423

RESUMO

The highly conserved herpesvirus glycoprotein complex gB/gH-gL mediates membrane fusion during virion entry and cell-cell fusion. Varicella-zoster virus (VZV) characteristically forms multinucleated cells, or syncytia, during the infection of human tissues, but little is known about this process. The cytoplasmic domain of VZV gB (gBcyt) has been implicated in cell-cell fusion regulation because a gB[Y881F] substitution causes hyperfusion. gBcyt regulation is necessary for VZV pathogenesis, as the hyperfusogenic mutant gB[Y881F] is severely attenuated in human skin xenografts. In this study, gBcyt-regulated fusion was investigated by comparing melanoma cells infected with wild-type-like VZV or hyperfusogenic mutants. The gB[Y881F] mutant exhibited dramatically accelerated syncytium formation in melanoma cells caused by fusion of infected cells with many uninfected cells, increased cytoskeleton reorganization, and rapid displacement of nuclei to dense central structures compared to pOka using live-cell confocal microscopy. VZV and human transcriptomes were concurrently investigated using whole transcriptome sequencing (RNA-seq) to identify viral and cellular responses induced when gBcyt regulation was disrupted by the gB[Y881F] substitution. The expression of four vital VZV genes, ORF61 and the genes for glycoproteins gC, gE, and gI, was significantly reduced at 36 h postinfection for the hyperfusogenic mutants. Importantly, hierarchical clustering demonstrated an association of differential gene expression with dysregulated gBcyt-mediated fusion. A subset of Ras GTPase genes linked to membrane remodeling were upregulated in cells infected with the hyperfusogenic mutants. These data implicate gBcyt in the regulation of gB fusion function that, if unmodulated, triggers cellular processes leading to hyperfusion that attenuates VZV infection. IMPORTANCE: The highly infectious, human-restricted pathogen varicella-zoster virus (VZV) causes chickenpox and shingles. Postherpetic neuralgia (PHN) is a common complication of shingles that manifests as prolonged excruciating pain, which has proven difficult to treat. The formation of fused multinucleated cells in ganglia might be associated with this condition. An effective vaccine against VZV is available but not recommended for immunocompromised individuals, highlighting the need for new therapies. This study investigated the viral and cellular responses to hyperfusion, a condition where the usual constraints of cell membranes are overcome and cells form multinucleated cells. This process hinders VZV and is regulated by a viral glycoprotein, gB. A combination of live-cell imaging and next-generation genomics revealed an alteration in viral and cellular responses during hyperfusion that was caused by the loss of gB regulation. These studies reveal mechanisms central to VZV pathogenesis, potentially leading to improved therapies.


Assuntos
Herpesvirus Humano 3/genética , Interações Hospedeiro-Patógeno , Melanócitos/virologia , Transcriptoma , Proteínas do Envelope Viral/genética , Proteínas ras/genética , Substituição de Aminoácidos , Fusão Celular , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Ontologia Genética , Genes Reporter , Células Gigantes/imunologia , Células Gigantes/ultraestrutura , Células Gigantes/virologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Herpesvirus Humano 3/crescimento & desenvolvimento , Herpesvirus Humano 3/imunologia , Humanos , Melanócitos/imunologia , Melanócitos/ultraestrutura , Anotação de Sequência Molecular , Mutação , Domínios Proteicos , Análise de Sequência de RNA , Transdução de Sinais , Proteínas do Envelope Viral/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia , Internalização do Vírus , Proteínas ras/imunologia
3.
Cytokine ; 76(2): 572-576, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26159111

RESUMO

Melanocytes are melanin-producing cells and with emerging innate immune functions including the expression of antiviral interferon-type I cytokines. We herein ascertained the susceptibility of the human melanocytes to Ross River alphavirus (RRV) infection and analyzed the subsequent immune responses. We demonstrated for the first time that (1) SKMEL-28 melanocyte cell line was susceptible to RRV infection and displaying major cytopathic activities and (2) RRV interfered with the interferon-type I response by altering nuclear translocation of pSTAT1 and pSTAT2 in infected SKMEL-28. These results suggest that the human melanoma cell line SKMEL-28 is a valuable model to analyze the mechanisms involved in severe skin manifestations and melanocyte's immunity at the portal of entry of major infection by arboviruses.


Assuntos
Interferon Tipo I/genética , Melanócitos/metabolismo , Ross River virus/patogenicidade , Linhagem Celular , Efeito Citopatogênico Viral , Humanos , Melanócitos/virologia
4.
J Invest Dermatol ; 135(12): 3009-3016, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26203638

RESUMO

Herpes simplex virus type 1 (HSV-1) invades its human host via the skin or mucosa. We aim to understand how HSV-1 overcomes the barrier function of the host epithelia, and for this reason, we established an ex vivo infection assay initially with murine skin samples. Here, we report how tissue has to be prepared to be susceptible to HSV-1 infection. Most efficient infection of the epidermis was achieved by removing the dermis. HSV-1 initially invaded the basal epidermal layer, and from there, spreading to the suprabasal layers was observed. Strikingly, in resting stage hair follicles, only the hair germ was infected, whereas the quiescent bulge stem cells (SCs) were resistant to infection. However, during the growth phase, infected cells were also detected in the activated bulge SCs. We demonstrated that cell proliferation was not a precondition for HSV-1 invasion, but SC activation was required as shown by infection of aberrantly activated bulge SCs in integrin-linked kinase (ILK)-deficient hair follicles. These results suggest that the status of the bulge SCs determines whether HSV-1 can reach its receptors, whereas the receptors on basal keratinocytes are accessible irrespective of their proliferation status.


Assuntos
Epiderme/virologia , Herpesvirus Humano 1/patogenicidade , Animais , Ácido Edético/farmacologia , Endopeptidases/farmacologia , Folículo Piloso/virologia , Proteínas Imediatamente Precoces/fisiologia , Melanócitos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Junções Íntimas/fisiologia , Ubiquitina-Proteína Ligases/fisiologia
5.
Exp Dermatol ; 24(4): 298-300, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25515776

RESUMO

Considerable evidence implicates that viral infection might be a participant factor in the pathogenesis of vitiligo. However, it is still unclear how viral infection leads to the melanocyte destruction. To elucidate the effects of viral dsDNA on the viability and cytokine synthesis of normal human melanocytes and to explore the underlying mechanisms, primary cultured normal human melanocytes were transfected with poly(dA:dT). The results demonstrated that poly(dA:dT) triggered apoptosis instead of pyroptosis in melanocytes. Knocking down AIM2 or RIG-I by RNA interference partially reduced the poly(dA:dT)-induced LDH release, suggesting the involvement of both nucleic acid sensors in the process of melanocyte death. Poly(dA:dT) induced the expression of pro-inflammatory cytokine genes including IFN-ß, TNF-α, IL-6 and IL-8 as well, whereas the pro-inflammatory cytokine production was suppressed by RIG-I siRNA, but not by AIM2 siRNA. Poly(dA:dT) treatment increased the phosphorylation of p38 and JNK and NFκB. Accordingly, NFκB inhibitor Bay 11-7082 and JNK inhibitor SP600125 blocked the induction of the cytokine genes except IFN-ß. The production of IL6 and IL8 was also suppressed by p38 inhibitor SB203580. On the contrary, the Poly(dA:dT)-induced melanocyte death was only decreased by SP600125. This study provides the possible mechanism of melanocyte destruction and immuno-stimulation in vitiligo by innate immune response following viral infection.


Assuntos
DNA Viral/imunologia , Melanócitos/citologia , Melanócitos/imunologia , Apoptose , Células Cultivadas , Citocinas/biossíntese , Citosol/imunologia , Citosol/virologia , Humanos , Imunidade Inata , Mediadores da Inflamação/metabolismo , Sistema de Sinalização das MAP Quinases/imunologia , Melanócitos/virologia , NF-kappa B/imunologia , Poli dA-dT/imunologia , Viroses/complicações , Viroses/imunologia , Vitiligo/etiologia , Vitiligo/imunologia , Vitiligo/patologia
6.
J Virol ; 88(22): 13086-98, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25187554

RESUMO

UNLABELLED: Cancer cells are susceptible to oncolytic viruses, albeit variably. Human adenoviruses (HAdVs) are widely used oncolytic agents that have been engineered to produce progeny within the tumor and elicit bystander effects. We searched for host factors enhancing bystander effects and conducted a targeted RNA interference screen against guanine nucleotide exchange factors (GEFs) of small GTPases. We show that the unfolded protein response (UPR), which is readily inducible in aggressive tumor cells, enhances melanoma or epithelial cancer cell killing upon HAdV infection. UPR was triggered by knockdown of Golgi-specific brefeldin A-resistant guanine nucleotide exchange factor 1 (GBF-1) or the GBF-1 inhibitor golgicide A (GCA) and stimulated HAdV infection. GBF-1 is a GEF for ADP ribosylation factors (Arfs) regulating endoplasmic reticulum (ER)-to-Golgi apparatus and intra-Golgi apparatus membrane transport. Cells treated with GCA enhanced HAdV-induced cytopathic effects in epithelial and melanoma cancer cells but not normal cells, if the drug was applied several hours prior to HAdV inoculation. This was shown by real-time label-free impedance measurements using the xCELLigence system. GCA-treated cells contained fewer incoming HAdVs than control cells, but GCA treatment boosted HAdV titers and spreading in cancer cells. GCA enhanced viral gene expression or transgene expression from the cytomegalovirus promoter of B- or C-species HAdVs but did not enhance viral early region 1A (E1A) expression in uninfected cell lines or cells transfected with plasmid reporter DNA. The UPR-enhanced cell killing required the nuclease activity of the UPR sensor inositol-requiring enzyme 1 (IRE-1) and X box binding protein 1 (XBP-1), which alleviate ER stress. The collective results show that chemical UPR induction and viruses boost tumor cell killing by enhancing oncolytic viral efficacy. IMPORTANCE: Cancer is difficult to combat. A wide range of oncolytic viruses show promise for killing cancer cells, yet the efficacy of oncolytic killing is low. We searched for host factors enhancing adenovirus cancer cell killing and found that the knockdown of Golgi-specific brefeldin A-resistant guanine nucleotide exchange factor 1 (GBF-1) or chemical inhibition of GBF-1 enhanced adenovirus infection by triggering the IRE-1/XBP-1 branch of the unfolded protein response (UPR). IRE-1/XBP-1 promote cell survival and enhanced the levels of the adenoviral immediate early gene product E1A, virus spreading, and killing of cancer cells. Aggressive tumor cells depend on a readily inducible UPR and, hence, present prime targets for a combined strategy involving adenoviruses and small chemicals inducing UPR.


Assuntos
Morte Celular , Células Epiteliais/virologia , Melanócitos/virologia , Vírus Oncolíticos/crescimento & desenvolvimento , Resposta a Proteínas não Dobradas , Linhagem Celular Tumoral , Células Epiteliais/fisiologia , Humanos , Melanócitos/fisiologia
7.
J Virol ; 88(9): 4932-42, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24554651

RESUMO

UNLABELLED: Members of the genus Parvovirus are small, nonenveloped single-stranded DNA viruses that are nonpathogenic in humans but have potential utility as cancer therapeutics. Because the innate immune response to parvoviruses has received relatively little attention, we compared the response to parvoviruses to that of several other types of viruses in human cells. In normal human glia, fibroblasts, or melanocytes, vesicular stomatitis virus evoked robust beta interferon (IFN-ß) responses. Cytomegalovirus, pseudorabies virus, and Sindbis virus all evoked a 2-log-unit or greater upregulation of IFN-ß in glia; in contrast, LuIII and MVMp parvoviruses did not evoke a detectable IFN-ß or interferon-stimulated gene (ISG; MX1, oligoadenylate synthetase [OAS], IFIT-1) response in the same cell types. The lack of response raised the question of whether parvoviral infection can be attenuated by IFN; interestingly, we found that IFN did not decrease parvovirus (MVMp, LuIII, and H-1) infectivity in normal human glia, fibroblasts, or melanocytes. The same was true in human cancers, including glioma, sarcoma, and melanoma. Similarly, IFN failed to attenuate transduction by the dependovirus vector adeno-associated virus type 2. Progeny production of parvoviruses was also unimpaired by IFN in both glioma and melanoma, whereas vesicular stomatitis virus replication was blocked. Sarcoma cells with upregulated IFN signaling that show high levels of resistance to other viruses showed strong infection by LuIII. Unlike many other oncolytic viruses, we found no evidence that impairment of innate immunity in cancer cells plays a role in the oncoselectivity of parvoviruses in human cells. Parvoviral resistance to the effects of IFN in cancer cells may constitute an advantage in the virotherapy of some tumors. IMPORTANCE: Understanding the interactions between oncolytic viruses and the innate immune system will facilitate employing these viruses as therapeutic agents in cancer patients. The cancer-selective nature of some oncolytic viruses is based on the impaired innate immunity of many cancer cells. The parvoviruses H-1, LuIII, and MVM target cancer cells; however, their relationship with the innate immune system is relatively uncharacterized. Surprisingly, we found that these parvoviruses do not evoke an interferon response in normal human fibroblasts, glia, or melanocytes. Furthermore, unlike most other types of virus, we found that parvovirus infectivity is unaffected by interferon treatment of human normal or tumor cells. Finally, parvoviral replication was unimpaired by interferon in four human tumor types, including those with residual interferon functionality. We conclude that deficits in the interferon antiviral response of cancer cells do not contribute to parvoviral oncoselectivity in human cells. The interferon-resistant phenotype of parvoviruses may give them an advantage over interferon-sensitive oncolytic viruses in tumors showing residual interferon functionality.


Assuntos
Interferon Tipo I/imunologia , Parvovirus/imunologia , Linhagem Celular , Fibroblastos/imunologia , Fibroblastos/virologia , Perfilação da Expressão Gênica , Humanos , Melanócitos/imunologia , Melanócitos/virologia , Neuroglia/imunologia , Neuroglia/virologia
8.
J Virol ; 87(12): 6644-59, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23552414

RESUMO

Metastatic malignant melanoma remains one of the most therapeutically challenging forms of cancer. Here we test replication-competent vesicular stomatitis viruses (VSV) on 19 primary human melanoma samples and compare these infections with those of normal human melanocyte control cells. Even at a low viral concentration, we found a strong susceptibility to viral oncolysis in over 70% of melanomas. In contrast, melanocytes displayed strong resistance to virus infection and showed complete protection by interferon. Several recombinant VSVs were compared, and all infected and killed most melanomas with differences in the time course with increasing rates of melanoma infection, as follows: VSV-CT9-M51 < VSV-M51 < VSV-G/GFP < VSV-rp30. VSV-rp30 sequencing revealed 2 nonsynonymous mutations at codon positions P126 and L223, both of which appear to be required for the enhanced phenotype. VSV-rp30 showed effective targeting and infection of multiple subcutaneous and intracranial melanoma xenografts in SCID mice after tail vein virus application. Sequence analysis of mutations in the melanomas used revealed that BRAF but not NRAS gene mutation status was predictive for enhanced susceptibility to infection. In mouse melanoma models with specific induced gene mutations including mutations of the Braf, Pten, and Cdkn2a genes, viral infection correlated with the extent of malignant transformation. Similar to human melanocytes, mouse melanocytes resisted VSV-rp30 infection. This study confirms the general susceptibility of the majority of human melanoma types for VSV-mediated oncolysis.


Assuntos
Melanócitos/virologia , Melanoma/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/patogenicidade , Vírus da Estomatite Vesicular Indiana/patogenicidade , Animais , Linhagem Celular Tumoral , Humanos , Melanoma/virologia , Camundongos , Camundongos SCID , Mutação , Vírus Oncolíticos/genética , Proteínas Proto-Oncogênicas B-raf/genética , Transplante Heterólogo/fisiologia , Vírus da Estomatite Vesicular Indiana/genética , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Exp Cell Res ; 315(5): 849-62, 2009 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-19167380

RESUMO

Melanoma development is a multi-step process arising from a series of genetic and epigenetic events. Although the sequential stages involved in progression from melanocytes to malignant melanoma are clearly defined, our current understanding of the mechanisms leading to melanoma onset is still incomplete. Growing evidence show that the activation of endogenous retroviral sequences might be involved in transformation of melanocytes as well as in the increased ability of melanoma cells to escape immune surveillance. Here we show that human melanoma cells in vitro undergo a transition from adherent to a more malignant, non-adherent phenotype when exposed to stress conditions. Melanoma-derived non-adherent cells are characterized by an increased proliferative potential and a decreased expression of both HLA class I molecules and Melan-A/MART-1 antigen, similarly to highly malignant cells. These phenotypic and functional modifications are accompanied by the activation of human endogenous retrovirus K expression (HERV-K) and massive production of viral-like particles. Down-regulation of HERV-K expression by RNA interference prevents the transition from the adherent to the non-adherent growth phenotype in low serum. These results implicate HERV-K in at least some critical steps of melanoma progression.


Assuntos
Transformação Celular Viral , Retrovirus Endógenos/fisiologia , Melanoma/virologia , Ativação Viral/fisiologia , Células CACO-2 , Proliferação de Células , Transformação Celular Viral/genética , Células Cultivadas , Células Clonais/virologia , Progressão da Doença , Retrovirus Endógenos/genética , Humanos , Células Jurkat , Células K562 , Melanócitos/patologia , Melanócitos/ultraestrutura , Melanócitos/virologia , Melanoma/etiologia , Melanoma/genética , Melanoma/patologia , Modelos Biológicos , RNA Viral/isolamento & purificação , Vírion/crescimento & desenvolvimento , Ativação Viral/genética
10.
J Cutan Pathol ; 35(8): 782-8, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18430043

RESUMO

A 65-year-old Latino man presented to his dermatologist for the removal of two melanocytic nevi from the back. The first nevus was removed from the right scapula and contained melanocytes with prominent eosinophilic nuclear inclusion bodies. The second nevus was removed from the paravertebral region, without evidence of inclusion bodies. Ultrastructurally, the inclusions in the first nevus contained dispersed finely granular, homogenous bodies without a limiting membrane. Immunohistochemistry characterized them as ubiquitin-positive material. Reverse transcriptase in situ polymerase chain reaction analysis was positive for molluscum-specific primers, suggesting that the inclusions encountered in the first nevus were secondary to a remote, local molluscum viral infection of melanocytes.


Assuntos
Corpos de Inclusão Intranuclear/patologia , Corpos de Inclusão Intranuclear/virologia , Melanócitos/patologia , Molusco Contagioso/patologia , Nevo Pigmentado/patologia , Nevo Pigmentado/virologia , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/virologia , Idoso , Humanos , Corpos de Inclusão Intranuclear/metabolismo , Masculino , Melanócitos/metabolismo , Melanócitos/virologia , Molusco Contagioso/complicações , Molusco Contagioso/metabolismo , Vírus do Molusco Contagioso/metabolismo , Nevo Pigmentado/metabolismo , RNA Viral/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Cutâneas/metabolismo
11.
J Submicrosc Cytol Pathol ; 35(3): 315-21, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-14690181

RESUMO

After Greene melanoma tissue had been implanted subcutaneously in a Syrian Golden Hamster and allowed to grow there for 10 days, its tissue components were examined by transmission electron microscopy. Amelanotic and melanotic tumor cells, blood vessels and nerve fibers were present. In the amelanotic and melanotic tumor cells the rough endoplasmic reticulum was colonized by spherical endogenous retroviruses, whereas no virus particles could be detected in endothelial or other cells. The cell-type-specific viruses thus mark separate cell lineages and indicate that blood vessels in Greene melanoma are not formed by tumor cells, so that the 'vasculogenic mimicry' currently under controversial discussion for other melanomas is not involved here. The tumor also comprised smooth muscle cells, skeletal muscle fibers and collagen fibers. This remarkable finding implies that tumor growth involves not only the stimulation of angiogenesis and neurogenesis by various growth factors, but also the activation of other cells/tissues in the adjacent host tissues. This influence of the surrounding host tissue produces the observed epiphenomena and can also cause the genotypic character of a tumor to be phenotypically masked (mimicry).


Assuntos
Transformação Celular Neoplásica , Melanócitos/ultraestrutura , Melanoma Experimental/patologia , Neovascularização Patológica/patologia , Neoplasias Cutâneas/patologia , Animais , Cricetinae , Retículo Endoplasmático Rugoso/ultraestrutura , Endotélio Vascular/ultraestrutura , Endotélio Vascular/virologia , Melanócitos/virologia , Melanoma Experimental/irrigação sanguínea , Transplante de Neoplasias , Fenótipo , Retroviridae/isolamento & purificação , Retroviridae/ultraestrutura , Neoplasias Cutâneas/irrigação sanguínea
12.
Cell Biol Int ; 25(1): 91-102, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11237412

RESUMO

Transformation of mouse melanocytes with a variety of exogenous oncogenes or chemical carcinogens frequently results in irreversible loss of pigmentation. We have infected mouse melanocytes with a temperature-sensitive mutant of the simian virus 40 (SV40) large tumour antigen to study the molecular mechanisms underlying depigmentation during melanocyte transformation. The results show that, out of six cell lines analyzed at the permissive temperature of the oncoprotein, three epidermal and two dermal melanocyte clones remained pigmented and retained the ability to synthesize melanin and to express the melanocyte-specific genes, including tyrosinase, tyrosinase related protein-1, tyrosinase related protein-2 and Mitf. In contrast, one dermal melanocyte clone (DMEL-3) gradually depigmented. This depigmentation was characterized by enhanced growth and down-regulation of melanocyte-specific gene expression. When the oncogene was inactivated by culture at the non-permissive temperature, the pigmented phenotype in DMEL-3 cells could be rescued, and there was a corresponding time-dependent increase in melanocyte-specific gene expression. After extended passage, this rescue could not be achieved. Our results provide direct evidence for the role of the SV40 large T antigen in melanocyte de-differentiation. Expression of Pax-3, a transcription factor implicated in melanocyte differentiation, was unaltered during the SV40-initiated de-differentiation, and de-differentiated melanocytes expressed normal levels of Pax-3 message. We speculate on the mechanism by which the oncoprotein might be regulating Mitf gene expression and of the role of Pax-3 in this process.


Assuntos
Antígenos Transformantes de Poliomavirus/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Interferon Tipo I/antagonistas & inibidores , Oxirredutases Intramoleculares/antagonistas & inibidores , Melanócitos/metabolismo , Monofenol Mono-Oxigenase/antagonistas & inibidores , Proteínas da Gravidez/antagonistas & inibidores , Fatores de Transcrição , Células 3T3 , Animais , Northern Blotting , Southern Blotting , Western Blotting , Linhagem Celular , Células Cultivadas , Melaninas/biossíntese , Melaninas/metabolismo , Melanócitos/virologia , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Fator de Transcrição Associado à Microftalmia , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados , Pigmentação/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Pele/citologia , Temperatura
14.
Int J Cancer ; 76(3): 430-6, 1998 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-9579583

RESUMO

We reported previously that B16,JB/RH and JB/MS melanomas of C57BL/6 mice express the common melanoma-associated antigen (MAA) recognized by MM2-9B6 monoclonal antibody (MAb). This MAA is encoded by the env gene of an ecotropic MuLV-type retrovirus that somatically emerged in melanomas of C57BL/6 mice. The potential role of this melanoma-associated retrovirus (MelARV) in melanoma formation remains unknown and has not been previously investigated. To test this, normal melanocyte lines (melan-a and C57M) of C57BL/6 mice were infected with the MelARV produced by B16BL6 melanoma. Infection of these melanocytes with the MelARV was associated with the appearance of the MAA recognized by MM2-9B6 MAb. Most of the infected melanocyte sublines were able to grow only in the presence of 12-O-tetradecanoylphorbol-13-acetate (TPA). Two infected melanocyte sublines showed morphological changes, were able to grow in the absence of TPA and, after inoculation into C57BL/6 mice, produced rapidly growing, highly pigmented tumors. These new melanomas, derived from the MelARV-infected melan-a and C57M melanocytes, were termed Meli-A1 and Meli-BL, respectively. Southern blot analysis of EcoRI- and HindIII-digested DNAs from these melanomas showed several retroviral insertion sites. One copy of MeIARV was found to be inserted at the end of the 6th leucine domain of the c-maf proto-oncogene, which encodes a basic region/leucine zipper transcription factor related to the AP-1 family that is able to form homodimers or heterodimers with Fos and Jun transcription factors. Our data indicate that c-maf is a common insertion site of MelARV in BL6, Meli-A1 and Meli-BL melanomas, whereas no such insertion site was found in the melanocytes infected with MelARV but not malignantly transformed. Thus, our data imply that the ecotropic MelARV that somatically emerged in B16 and other melanomas of C57BL/6 mice may play a role in malignant transformation.


Assuntos
Transformação Celular Neoplásica , Gammaretrovirus/fisiologia , Melanócitos/virologia , Melanoma Experimental/virologia , Provírus/fisiologia , Animais , Transformação Celular Viral , Gammaretrovirus/isolamento & purificação , Gammaretrovirus/ultraestrutura , Melanócitos/patologia , Melanócitos/fisiologia , Melanoma Experimental/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica , Provírus/isolamento & purificação , Mapeamento por Restrição
15.
J Virol ; 69(8): 4994-5010, 1995 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7609070

RESUMO

The pathway of envelopment and egress of the varicella-zoster virus (VZV) and the primary site of viral production within the epidermal layer of the skin are not fully understood. There are several hypotheses to explain how the virus may receive an envelope as it travels to the surface of the monolayer. In this study, we expand earlier reports and provide a more detailed explanation of the growth of VZV in human melanoma cells. Human melanoma cells were selected because they are a malignant derivative of the melanocyte, the melanin-producing cell which originates in the neural crest. We were able to observe the cytopathic effects of syncytial formation and the pattern of egress of virions at the surfaces of infected monolayers by scanning electron microscopy and laser-scanning confocal microscopy. The egressed virions did not appear uniformly over the syncytial surface, rather they were present in elongated patterns which were designated viral highways. In order to document the pathway by which VZV travels from the host cell nucleus to the outer cell membrane, melanoma cells were infected and then processed for examination by transmission electron microscopy (TEM) at increasing intervals postinfection. At the early time points, within minutes to hours postinfection, it was not possible to localize the input virus by TEM. Thus, viral particles first observed at 24 h postinfection were considered progeny virus. On the basis of the TEM observations, the following sequence of events was considered most likely. Nucleocapsids passed through the inner nuclear membrane and acquired an envelope, after which they were seen in the endoplasmic reticulum. Enveloped virions within vacuoles derived from the endoplasmic reticulum passed into the cytoplasm. Thereafter, vacuoles containing nascent enveloped particles acquired viral glycoproteins by fusion with vesicles derived from the Golgi. The vacuoles containing virions fused with the outer plasma membrane and the particles appeared on the surface of the infected cell. Late in infection, enveloped virions were also present within the nuclei of infected cells; the most likely mechanism was retrograde flow from the perinuclear space back into the nucleus. Thus, this study suggests a role for the melanocyte in the pathogenesis of VZV infection, because all steps in viral egress can be accounted for if VZV subsumes the cellular pathways required for melanogenesis.


Assuntos
Herpesvirus Humano 3/fisiologia , Melanócitos/virologia , Melanoma/virologia , Capsídeo/metabolismo , Núcleo Celular/ultraestrutura , Citoplasma/virologia , Células Gigantes/ultraestrutura , Células Gigantes/virologia , Herpesvirus Humano 3/ultraestrutura , Humanos , Pulmão/virologia , Microscopia Eletrônica de Varredura , Células Tumorais Cultivadas , Vacúolos/virologia , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...