Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
1.
Cell ; 179(3): 632-643.e12, 2019 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-31607510

RESUMO

Antisense Piwi-interacting RNAs (piRNAs) guide silencing of established transposons during germline development, and sense piRNAs drive ping-pong amplification of the antisense pool, but how the germline responds to genome invasion is not understood. The KoRV-A gammaretrovirus infects the soma and germline and is sweeping through wild koalas by a combination of horizontal and vertical transfer, allowing direct analysis of retroviral invasion of the germline genome. Gammaretroviruses produce spliced Env mRNAs and unspliced transcripts encoding Gag, Pol, and the viral genome, but KoRV-A piRNAs are almost exclusively derived from unspliced genomic transcripts and are strongly sense-strand biased. Significantly, selective piRNA processing of unspliced proviral transcripts is conserved from insects to placental mammals. We speculate that bypassed splicing generates a conserved molecular pattern that directs proviral genomic transcripts to the piRNA biogenesis machinery and that this "innate" piRNA response suppresses transposition until antisense piRNAs are produced, establishing sequence-specific adaptive immunity.


Assuntos
Gammaretrovirus/genética , Phascolarctidae/genética , RNA Interferente Pequeno/genética , Animais , Elementos de DNA Transponíveis , Gammaretrovirus/metabolismo , Gammaretrovirus/patogenicidade , Produtos do Gene env/genética , Produtos do Gene env/metabolismo , Produtos do Gene gag/genética , Produtos do Gene gag/metabolismo , Produtos do Gene pol/genética , Produtos do Gene pol/metabolismo , Genoma , Células Germinativas/metabolismo , Células Germinativas/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Phascolarctidae/virologia , Splicing de RNA , RNA Antissenso/genética , RNA Antissenso/metabolismo , RNA Interferente Pequeno/metabolismo
2.
J Virol ; 93(11)2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30894464

RESUMO

Viruses can incorporate foreign glycoproteins to form infectious particles through a process known as pseudotyping. However, not all glycoproteins are compatible with all viruses. Despite the fact that viral pseudotyping is widely used, what makes a virus/glycoprotein pair compatible is poorly understood. To study this, we chose to analyze a gammaretroviral glycoprotein (Env) whose compatibility with different viruses could be modulated through small changes in its cytoplasmic tail (CT). One form of this glycoprotein is compatible with murine leukemia virus (MLV) particles but incompatible with human immunodeficiency virus type 1 (HIV-1) particles, while the second is compatible with HIV-1 particles but not with MLV particles. To decipher the factors affecting virus-specific Env incompatibility, we characterized Env incorporation, maturation, cell-to-cell fusogenicity, and virus-to-cell fusogenicity of each Env. The HIV-1 particle incompatibility correlated with less efficient cleavage of the R peptide by HIV-1 protease. However, the MLV particle incompatibility was more nuanced. MLV incompatibility appeared to be caused by lack of incorporation into particles, yet incorporation could be restored by further truncating the CT or by using a chimeric MLV Gag protein containing the HIV-1 MA without fully restoring infectivity. The MLV particle incompatibility appeared to be caused in part by fusogenic repression in MLV particles through an unknown mechanism. This study demonstrates that the Env CT can dictate functionality of Env within particles in a virus-specific manner.IMPORTANCE Viruses utilize viral glycoproteins to efficiently enter target cells during infection. How viruses acquire viral glycoproteins has been studied to understand the pathogenesis of viruses and develop safer and more efficient viral vectors for gene therapies. The CTs of viral glycoproteins have been shown to regulate various stages of glycoprotein biogenesis, but a gap still remains in understanding the molecular mechanism of glycoprotein acquisition and functionality regarding the CT. Here, we studied the mechanism of how specific mutations in the CT of a gammaretroviral envelope glycoprotein distinctly affect infectivity of two different viruses. Different mutations caused failure of glycoproteins to function in a virus-specific manner due to distinct fusion defects, suggesting that there are virus-specific characteristics affecting glycoprotein functionality.


Assuntos
Gammaretrovirus/genética , Produtos do Gene env/genética , Proteínas do Envelope Viral/genética , Animais , Linhagem Celular , Membrana Celular/metabolismo , Gammaretrovirus/metabolismo , Produtos do Gene env/metabolismo , Produtos do Gene gag/genética , Células HEK293 , HIV-1/metabolismo , Humanos , Vírus da Leucemia Murina/genética , Vírus da Leucemia Murina/metabolismo , Camundongos , Mutação , Especificidade da Espécie , Vírion/metabolismo , Montagem de Vírus
3.
J Virol ; 93(5)2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30541852

RESUMO

Approximately 10% of the mouse genome is composed of endogenous retroviruses belonging to different families. In contrast to the situation in the human genome, several of these families correspond to recent, still-infectious elements capable of encoding complete viral particles. The mouse GLN endogenous retrovirus is one of these active families. We previously identified one fully functional provirus from the sequenced genome of the C57BL/6 mouse strain. The GLN envelope protein gives the infectious viral particles an ecotropic host range, and we had demonstrated that the receptor was neither CAT1 nor SMIT1, the two previously identified receptors for mouse ecotropic retroviral envelope proteins. In this study, we have identified SLC19A1, the reduced folate carrier, as the cellular protein used as a receptor by the GLN retrovirus. The ecotropic tropism exhibited by this envelope is due to the presence or absence of an N-linked glycosylation site in the first extracellular loop as well as the specific amino acid sequence of the extracellular domains of the receptor. Like all the other retroviral envelope proteins from the gammaretrovirus genus whose receptors have been identified, the GLN envelope protein uses a member of the solute carrier superfamily as a receptor.IMPORTANCE Endogenous retroviruses are genomic traces of past infections present in all vertebrates. Most of these elements degenerate over time and become nonfunctional, but the mouse genome still contains several families with full infection abilities. The GLN retrovirus is one of them, and its members encode particles that are able to infect only mouse cells. Here, we identified the cellular protein used as a receptor by GLN for cell entry. It is SLC19A1, the reduced folate carrier. We show that GLN infection is limited to mouse cells due to both a mutation in the mouse gene preventing the glycosylation of SLC19A1 and also other residues conserved within the rat but not in the hamster and human proteins. Like all other gammaretroviruses whose receptors have been identified, GLN uses a member of the solute carrier superfamily for cell entry, highlighting the role of these proteins for retroviral infection in mammals.


Assuntos
Gammaretrovirus/metabolismo , Produtos do Gene env/genética , Receptores Virais/genética , Proteína Carregadora de Folato Reduzido/genética , Proteínas do Envelope Viral/genética , Ligação Viral , Animais , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Gammaretrovirus/genética , Genoma/genética , Glicosilação , Células HEK293 , Especificidade de Hospedeiro , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Proteína Carregadora de Folato Reduzido/metabolismo , Infecções por Retroviridae/virologia
4.
Cell Rep ; 21(1): 17-26, 2017 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-28978471

RESUMO

Antigen-independent tonic signaling by chimeric antigen receptors (CARs) can increase differentiation and exhaustion of T cells, limiting their potency. Incorporating 4-1BB costimulation in CARs may enable T cells to resist this functional exhaustion; however, the potential ramifications of tonic 4-1BB signaling in CAR T cells remain unclear. Here, we found that tonic CAR-derived 4-1BB signaling can produce toxicity in T cells via continuous TRAF2-dependent activation of the nuclear factor κB (NF-κB) pathway and augmented FAS-dependent cell death. This mechanism was amplified in a non-self-inactivating gammaretroviral vector through positive feedback on the long terminal repeat (LTR) promoter, further enhancing CAR expression and tonic signaling. Attenuating CAR expression by substitution with a self-inactivating lentiviral vector minimized tonic signaling and improved T cell expansion and anti-tumor function. These studies illuminate the interaction between tonic CAR signaling and the chosen expression platform and identify inhibitory properties of the 4-1BB costimulatory domain that have direct implications for rational CAR design.


Assuntos
Ligante 4-1BB/genética , Antígenos de Neoplasias/genética , Regulação Leucêmica da Expressão Gênica , Leucemia-Linfoma de Células T do Adulto/genética , Proteínas Mutantes Quiméricas/genética , Linfócitos T/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Ligante 4-1BB/imunologia , Animais , Antígenos de Neoplasias/imunologia , Morte Celular , Sobrevivência Celular , Gammaretrovirus/genética , Gammaretrovirus/metabolismo , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Leucemia-Linfoma de Células T do Adulto/imunologia , Leucemia-Linfoma de Células T do Adulto/patologia , Camundongos , Camundongos Endogâmicos NOD , Proteínas Mutantes Quiméricas/imunologia , NF-kappa B/genética , NF-kappa B/imunologia , Transplante de Neoplasias , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Transdução de Sinais , Linfócitos T/patologia , Linfócitos T/transplante , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Receptor fas/genética , Receptor fas/imunologia
5.
Virology ; 507: 140-150, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28437635

RESUMO

Porcine endogenous retrovirus-A (PERV-A), a gammaretrovirus, infects human cells in vitro, thus raising the potential risk of cross-species transmission in xenotransplantation. Two members of the solute carrier family 52 (SLC52A1 and SLC52A2) are PERV-A receptors. Site-directed mutagenesis of the cDNA encoding SLC52A1 identified that only one of two putative glycosylation signals is occupied by glycans. In addition, we showed that glycosylation of SLC52A1 is not necessary for PERV-A receptor function. We also identified that at a minimum, three cysteine residues are sufficient for SLC52A1 cell surface expression. Mutation of cysteine at position 365 and either of the two cysteine residues in the C-terminal tail at positions 442 or 446 reduced SLC52A1 surface expression and PERV-A infection suggesting that these residues may contribute to overall structural stability and receptor function. Understanding interactions between PERV-A and its cellular receptor may provide novel strategies to prevent zoonotic infection in the setting of xenotransplantation.


Assuntos
Cisteína/metabolismo , Retrovirus Endógenos/patogenicidade , Gammaretrovirus/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Virais/química , Receptores Virais/metabolismo , Infecções por Retroviridae/veterinária , Doenças dos Suínos/metabolismo , Animais , Cisteína/química , Cisteína/genética , Retrovirus Endógenos/genética , Retrovirus Endógenos/fisiologia , Gammaretrovirus/classificação , Gammaretrovirus/genética , Glicosilação , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Virais/genética , Infecções por Retroviridae/genética , Infecções por Retroviridae/metabolismo , Infecções por Retroviridae/virologia , Suínos , Doenças dos Suínos/genética , Doenças dos Suínos/virologia , Virulência
6.
Hum Gene Ther Clin Dev ; 25(4): 218-28, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25381930

RESUMO

The clinical application of self-inactivating (SIN) retroviral vectors requires an efficient vector production technology. To enable production of γ-retroviral SIN vectors from stable producer cells, new targetable HEK293-based producer clones were selected, providing amphotropic, GALV, or RD114 pseudotyping. Viral vector expression constructs can reliably be inserted at a predefined genomic locus via Flp-recombinase-mediated cassette exchange. Introduction of a clean-up step, mediated by Cre-recombinase, allows the removal of residual sequences that were required for targeting and selection, but were dispensable for the final producer clones and eliminated homology-driven recombination between the tagging and the therapeutic vector. The system was used to establish GALV and RD114 pseudotyping producer cells (HG- and HR820) for a clinically relevant long terminal repeat-driven therapeutic vector, designed for the transfer of a recombinant TCR that delivered titers in the range of 2×10(7) infectious particles (IP)/ml. Production capacity of the amphotropic producer cell (HA820) was challenged by a therapeutic SIN vector transferring the large COL7A1 cDNA. The final producer clone delivered a titer of 4×10(6) IP/ml and the vector containing supernatant was used directly to functionally restore primary fibroblasts and keratinocytes isolated from recessive dystrophic epidermolysis bullosa patients. Thus, the combinatorial approach (fc-technology) to generate producer cells for therapeutic γ-retroviral (SIN) vectors is feasible, is highly efficient, and allows their safe production and application in clinical trials.


Assuntos
Colágeno Tipo VII/genética , DNA Recombinante/genética , Gammaretrovirus/genética , Engenharia Genética/métodos , Vetores Genéticos/genética , Colágeno Tipo VII/metabolismo , DNA Recombinante/isolamento & purificação , Gammaretrovirus/metabolismo , Marcação de Genes/métodos , Vetores Genéticos/isolamento & purificação , Células HEK293 , Humanos
7.
Nucleic Acid Ther ; 24(5): 356-63, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25019196

RESUMO

To generate γ-retroviral vectors for stable conjoint expression of artificial microRNAs (amiR) and therapeutic genes in primary human lymphocytes, and to identify the design parameters that are key for successful vector generation. Gamma-retroviral vectors were designed to co-express both amiRs and a linked reporter gene, truncated CD34 (tCD34). Artificial miRs based on microRNAs miR-16, miR-142, miR-146b, miR-150, miR155, and miR-223 were inserted into sites within the intron of the vector and tested for tCD34 expression by flow cytometry (FACS). Different constructs were assembled with amiRs targeted to knockdown expression of suppressor of cytokine signaling 1 (SOCS1) or programmed cell death 1 (PDCD1, PD-1). Three of the six amiRs maintained tCD34 expression. Expansion of primary human T cells transduced with these amiR vectors, as well as transgene expression, were equivalent to control engineered T cells over a 40-day period. Knockdown of SOCS1 RNA and PD-1 expression by FACS was shown to vary between constructs, dependent on either the specific short interfering RNA sequence used in the amiR, or the microRNA backbone and location in the vector intron. Gamma-retroviral vectors that both efficiently knockdown endogenous gene expression and maintain linked transgene production can be produced, but empirical vector evaluations were best suited for optimal construct analysis.


Assuntos
Gammaretrovirus/genética , Vetores Genéticos/química , MicroRNAs/genética , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Proteínas Supressoras da Sinalização de Citocina/antagonistas & inibidores , Antígenos CD34/genética , Antígenos CD34/metabolismo , Proliferação de Células , Citometria de Fluxo , Gammaretrovirus/química , Gammaretrovirus/metabolismo , Expressão Gênica , Genes Reporter , Vetores Genéticos/metabolismo , Humanos , Íntrons , MicroRNAs/síntese química , MicroRNAs/metabolismo , Cultura Primária de Células , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/metabolismo , Relação Estrutura-Atividade , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Linfócitos T/citologia , Linfócitos T/metabolismo
8.
PLoS One ; 8(2): e56029, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23418499

RESUMO

To investigate mechanisms and phenotypic effects of insertional mutagenesis by gammaretroviruses, we have developed mouse lines containing a single Akv 1-99 long terminal repeat (LTR) and a floxed PGK/Tn5 neomycin cassette at the Nras proto-oncogene at positions previously identified as viral integration sites in Akv 1-99 induced tumors. The insert did not compromise the embryonic development, however, the cassette had an effect on Nras expression in all tissues analyzed. Cre-mediated excision of the PGK/Tn5 neomycin cassette in two of the lines caused upregulation of Nras. Altogether, the knock-in alleles are characterized by modulation of expression of the target gene from more than ten-fold upregulation to three-fold downregulation and exemplify various mechanisms of deregulation by insertional mutagenesis. LTR knock-in mice may serve as a tool to investigate mechanisms of retroviral insertional mutagenesis and as a way of constitutive or induced modulation of expression of a target gene.


Assuntos
Gammaretrovirus/genética , Genes ras/genética , Sequências Repetidas Terminais , Integração Viral , Alelos , Animais , Gammaretrovirus/metabolismo , Camundongos , Mutagênese Insercional
9.
Future Microbiol ; 8(1): 107-21, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23252496

RESUMO

Gene- and cell-based therapies hold great potential for the advancement of the personalized medicine movement. Gene therapy vectors have made dramatic leaps forward since their inception. Retroviral-based vectors were the first to gain clinical attention and still offer the best hope for the long-term correction of many disorders. The fear of nonspecific transduction makes targeting a necessary feature for most clinical applications. However, this remains a difficult feature to optimize, with specificity often coming at the expense of efficiency. The aim of this article is to discuss the various methods employed to retarget retroviral entry. Our focus will lie on the modification of gammaretroviral envelope proteins with an in-depth discussion of the creation and screening of envelope libraries.


Assuntos
Gammaretrovirus/genética , Gammaretrovirus/metabolismo , Vetores Genéticos/genética , Receptores Virais/metabolismo , Animais , Biblioteca Gênica , Humanos , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus
10.
J Virol ; 86(19): 10621-7, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22811542

RESUMO

We have constructed a replication-competent gammaretrovirus (SL3-AP) capable of using the human G-protein-coupled receptor hAPJ as its entry receptor. The envelope protein of the virus was made by insertion of the 13-amino-acid peptide ligand for hAPJ, flanked by linker sequences, into one of the variable loops of the receptor binding domain of SL3-2, a murine leukemia virus (MLV) that uses the xenotropic-polytropic virus receptor Xpr1 and which has a host range limited to murine cells. This envelope protein can utilize hAPJ as well as murine Xpr1 for entry into host cells with equal efficiencies. In addition, the SL3-AP virus replicates in cells expressing either of its receptors, hAPJ and murine Xpr1, and causes resistance to superinfection and downregulation of hAPJ in infected cells. Thus, SL3-AP is the first example of a retargeted replication-competent retrovirus, with replication characteristics and receptor interference properties similar to those of natural isolates.


Assuntos
Gammaretrovirus/metabolismo , Regulação Viral da Expressão Gênica , Receptores Acoplados a Proteínas G/fisiologia , Sequência de Aminoácidos , Animais , Receptores de Apelina , Citometria de Fluxo/métodos , Células HEK293 , Humanos , Cinética , Vírus da Leucemia Murina/metabolismo , Camundongos , Modelos Moleculares , Conformação Molecular , Dados de Sequência Molecular , Células NIH 3T3 , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Receptores Virais/metabolismo , Homologia de Sequência de Aminoácidos , Receptor do Retrovírus Politrópico e Xenotrópico
11.
BMC Biochem ; 12: 21, 2011 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-21586110

RESUMO

BACKGROUND: The inorganic (Pi) phosphate transporter (PiT) family comprises known and putative Na(+)- or H(+)-dependent Pi-transporting proteins with representatives from all kingdoms. The mammalian members are placed in the outer cell membranes and suggested to supply cells with Pi to maintain house-keeping functions. Alignment of protein sequences representing PiT family members from all kingdoms reveals the presence of conserved amino acids and that bacterial phosphate permeases and putative phosphate permeases from archaea lack substantial parts of the protein sequence when compared to the mammalian PiT family members. Besides being Na(+)-dependent P(i) (NaP(i)) transporters, the mammalian PiT paralogs, PiT1 and PiT2, also are receptors for gamma-retroviruses. We have here exploited the dual-function of PiT1 and PiT2 to study the structure-function relationship of PiT proteins. RESULTS: We show that the human PiT2 histidine, H(502), and the human PiT1 glutamate, E(70),--both conserved in eukaryotic PiT family members--are critical for P(i) transport function. Noticeably, human PiT2 H(502) is located in the C-terminal PiT family signature sequence, and human PiT1 E(70) is located in ProDom domains characteristic for all PiT family members.A human PiT2 truncation mutant, which consists of the predicted 10 transmembrane (TM) domain backbone without a large intracellular domain (human PiT2ΔR(254)-V(483)), was found to be a fully functional P(i) transporter. Further truncation of the human PiT2 protein by additional removal of two predicted TM domains together with the large intracellular domain created a mutant that resembles a bacterial phosphate permease and an archaeal putative phosphate permease. This human PiT2 truncation mutant (human PiT2ΔL(183)-V(483)) did also support P(i) transport albeit at very low levels. CONCLUSIONS: The results suggest that the overall structure of the P(i)-transporting unit of the PiT family proteins has remained unchanged during evolution. Moreover, in combination, our studies of the gene structure of the human PiT1 and PiT2 genes (SLC20A1 and SLC20A2, respectively) and alignment of protein sequences of PiT family members from all kingdoms, along with the studies of the dual functions of the human PiT paralogs show that these proteins are excellent as models for studying the evolution of a protein's structure-function relationship.


Assuntos
Biologia Computacional , Sequência Conservada , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/química , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/metabolismo , Sequência de Aminoácidos , Animais , Transporte Biológico , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Cães , Evolução Molecular , Éxons/genética , Gammaretrovirus/metabolismo , Humanos , Espaço Intracelular/metabolismo , Íntrons/genética , Camundongos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Alinhamento de Sequência , Deleção de Sequência , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/genética , Especificidade da Espécie
12.
Stem Cells Dev ; 20(5): 795-807, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21058905

RESUMO

Induced pluripotent stem (iPS) cells have great potential for regenerative medicine and gene therapy. Thus far, iPS cells have typically been generated using integrating viral vectors expressing various reprogramming transcription factors; nonintegrating methods have been less effective and efficient. Because there is a significant risk of malignant transformation and cancer involved with the use of iPS cells, careful evaluation of transplanted iPS cells will be necessary in small and large animal studies before clinical application. Here, we have generated and characterized nonhuman primate iPS cells with the goal of evaluating iPS cell transplantation in a clinically relevant large animal model. We developed stable Phoenix-RD114-based packaging cell lines that produce OCT4, SOX2, c-MYC, and KLF4 (OSCK) expressing gammaretroviral vectors. Using these vectors in combination with small molecules, we were able to efficiently and reproducibly generate nonhuman primate iPS cells from pigtailed macaques (Macaca nemestrina). The established nonhuman primate iPS cells exhibited pluripotency and extensive self-renewal capacity. The facile and reproducible generation of nonhuman primate iPS cells using defined producer cells as a source of individual reprogramming factors should provide an important resource to optimize and evaluate iPS cell technology for studies involving stem cell biology and regenerative medicine.


Assuntos
Reprogramação Celular , Fibroblastos/metabolismo , Células-Tronco Pluripotentes Induzidas , Macaca nemestrina/genética , Animais , Diferenciação Celular/genética , Linhagem Celular , Reprogramação Celular/genética , Feminino , Fibroblastos/citologia , Gammaretrovirus/genética , Gammaretrovirus/metabolismo , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Macaca nemestrina/metabolismo , Masculino , Camundongos , Análise em Microsséries , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Medicina Regenerativa , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Risco , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Transfecção
13.
PLoS One ; 5(7): e11738, 2010 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-20668529

RESUMO

BACKGROUND: The human exogenous gammaretrovirus XMRV is thought to be implicated in prostate cancer and chronic fatigue syndrome. Besides pressing epidemiologic questions, the elucidation of the tissue and cell tropism of the virus, as well as its sensitivity to retroviral restriction factors is of fundamental importance. The Apobec3 (A3) proteins, a family of cytidine deaminases, are one important group of host proteins that control primary infection and efficient viral spread. METHODOLOGY/PRINCIPAL FINDINGS: Here we demonstrate that XMRV is resistant to human Apobec 3B, 3C and 3F, while being highly susceptible to the human A3G protein, a factor which is known to confer antiviral activity against most retroviruses. We show that XMRV as well as MoMLV virions package Apobec proteins independent of their specific restriction activity. hA3G was found to be a potent inhibitor of XMRV as well as of MoMLV infectivity. In contrast to MoMLV, XMRV infection can also be partially reduced by low concentrations of mA3. Interestingly, established prostate cancer cell lines, which are highly susceptible to XMRV infection, do not or only weakly express hA3G. CONCLUSIONS: Our findings confirm and extend recently published data that show restriction of XMRV infection by hA3G. The results will be of value to explore which cells are infected with XMRV and efficiently support viral spread in vivo. Furthermore, the observation that XMRV infection can be reduced by mA3 is of interest with regard to the current natural reservoir of XMRV infection.


Assuntos
Citidina Desaminase/metabolismo , Citosina Desaminase/metabolismo , Gammaretrovirus/metabolismo , Desaminases APOBEC , Desaminase APOBEC-3G , Animais , Linhagem Celular , Citidina Desaminase/genética , Citosina Desaminase/genética , Gammaretrovirus/genética , Vetores Genéticos/genética , Humanos , Immunoblotting , Camundongos , Antígenos de Histocompatibilidade Menor , Reação em Cadeia da Polimerase , RNA Mensageiro , Retroviridae/genética , Células Tumorais Cultivadas , Vírion/genética
14.
J Virol ; 84(5): 2556-62, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20015990

RESUMO

Xenotropic murine leukemia virus-related virus (XMRV) is a novel human gammaretrovirus discovered in association with human prostate tumors. XMRV was first identified in prostate stromal cells surrounding the tumors of patients carrying a mutation in the HPC1 gene locus. To determine the tropism of XMRV in cell culture, we tested the ability of XMRV to spread and replicate in various prostate and nonprostate cell lines. We found that although the expression of XMRV viral proteins and the spread of infectious virus were minimal in a variety of cell lines, XMRV displayed robust expression and infection in LNCaP prostate tumor cells. The transcriptional activity of the XMRV long terminal repeat (LTR) was found to be higher than the Moloney murine leukemia virus LTRs in both LNCaP and WPMY-1 (simian virus 40-transformed prostate stromal cells). The U3 promoter of XMRV and a glucocorticoid response element (GRE) within the U3 were required for the transcriptional activity in LNCaP cells. Coexpression of the androgen receptor and stimulation with dihydrotestosterone stimulated XMRV-LTR-dependent transcription in 293T cells, and the GRE was required for this activity. These data suggest that XMRV may replicate more efficiently in LNCaP cells in part due to the transcriptional environment in LNCaP cells.


Assuntos
Gammaretrovirus , Próstata , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/virologia , Transcrição Gênica , Replicação Viral/fisiologia , Animais , Linhagem Celular Tumoral , Gammaretrovirus/genética , Gammaretrovirus/metabolismo , Gammaretrovirus/patogenicidade , Genes Reporter , Humanos , Masculino , Camundongos , Vírus da Leucemia Murina de Moloney/genética , Vírus da Leucemia Murina de Moloney/metabolismo , Regiões Promotoras Genéticas , Próstata/citologia , Próstata/patologia , Próstata/virologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Internalização do Vírus
15.
Proc Natl Acad Sci U S A ; 106(38): 16351-6, 2009 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-19805305

RESUMO

Xenotropic murine leukemia virus-related virus (XMRV) was recently discovered in human prostate cancers and is the first gammaretrovirus known to infect humans. While gammaretroviruses have well-characterized oncogenic effects in animals, they have not been shown to cause human cancers. We provide experimental evidence that XMRV is indeed a gammaretrovirus with protein composition and particle ultrastructure highly similar to Moloney murine leukemia virus (MoMLV), another gammaretrovirus. We analyzed 334 consecutive prostate resection specimens, using a quantitative PCR assay and immunohistochemistry (IHC) with an anti-XMRV specific antiserum. We found XMRV DNA in 6% and XMRV protein expression in 23% of prostate cancers. XMRV proteins were expressed primarily in malignant epithelial cells, suggesting that retroviral infection may be directly linked to tumorigenesis. XMRV infection was associated with prostate cancer, especially higher-grade cancers. We found XMRV infection to be independent of a common polymorphism in the RNASEL gene, unlike results previously reported. This finding increases the population at risk for XMRV infection from only those homozygous for the RNASEL variant to all individuals. Our observations provide evidence for an association of XMRV with malignant cells and with more aggressive tumors.


Assuntos
Gammaretrovirus/fisiologia , Próstata/virologia , Neoplasias da Próstata/virologia , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/virologia , Adulto , Idoso , Sequência de Bases , Western Blotting , DNA Viral/genética , Endorribonucleases/genética , Epitélio/patologia , Epitélio/virologia , Gammaretrovirus/genética , Gammaretrovirus/metabolismo , Genótipo , Interações Hospedeiro-Patógeno , Humanos , Imuno-Histoquímica , Masculino , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Próstata/patologia , Neoplasias da Próstata/patologia , Infecções por Retroviridae/patologia , Homologia de Sequência do Ácido Nucleico , Infecções Tumorais por Vírus/patologia , Carga Viral , Proteínas Virais/metabolismo , Vírion/genética , Vírion/imunologia , Vírion/ultraestrutura
16.
J Clin Invest ; 118(9): 3132-42, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18688285

RESUMO

Previously, several individuals with X-linked SCID (SCID-X1) were treated by gene therapy to restore the missing IL-2 receptor gamma (IL2RG) gene to CD34+ BM precursor cells using gammaretroviral vectors. While 9 of 10 patients were successfully treated, 4 of the 9 developed T cell leukemia 31-68 months after gene therapy. In 2 of these cases, blast cells contained activating vector insertions near the LIM domain-only 2 (LMO2) proto-oncogene. Here, we report data on the 2 most recent adverse events, which occurred in patients 7 and 10. In patient 10, blast cells contained an integrated vector near LMO2 and a second integrated vector near the proto-oncogene BMI1. In patient 7, blast cells contained an integrated vector near a third proto-oncogene,CCND2. Additional genetic abnormalities in the patients' blast cells included chromosomal translocations, gain-of-function mutations activating NOTCH1, and copy number changes, including deletion of tumor suppressor gene CDKN2A, 6q interstitial losses, and SIL-TAL1 rearrangement. These findings functionally specify a genetic network that controls growth in T cell progenitors. Chemotherapy led to sustained remission in 3 of the 4 cases of T cell leukemia, but failed in the fourth. Successful chemotherapy was associated with restoration of polyclonal transduced T cell populations. As a result, the treated patients continued to benefit from therapeutic gene transfer.


Assuntos
Cromossomos Humanos X , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Leucemia de Células T/etiologia , Imunodeficiência Combinada Severa/terapia , Proteínas Adaptadoras de Transdução de Sinal , Antineoplásicos/farmacologia , Aberrações Cromossômicas , Ciclina D2 , Ciclinas/genética , Proteínas de Ligação a DNA/genética , Gammaretrovirus/metabolismo , Humanos , Lactente , Janus Quinase 3/genética , Proteínas com Domínio LIM , Leucemia de Células T/complicações , Leucemia de Células T/terapia , Metaloproteínas/genética , Modelos Biológicos , Mutação , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas , Receptores de Interleucina-2/genética , Imunodeficiência Combinada Severa/complicações
17.
J Virol ; 82(20): 9964-77, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18684813

RESUMO

Xenotropic murine leukemia virus-related virus (XMRV) is a new human gammaretrovirus identified in prostate cancer tissue from patients homozygous for a reduced-activity variant of the antiviral enzyme RNase L. Neither a casual relationship between XMRV infection and prostate cancer nor a mechanism of tumorigenesis has been established. To determine the integration site preferences of XMRV and the potential risk of proviral insertional mutagenesis, we carried out a genome-wide analysis of viral integration sites in the prostate cell line DU145 after an acute XMRV infection and compared the integration site pattern of XMRV with those found for murine leukemia virus and two human retroviruses, human immunodeficiency virus type 1 and human T-cell leukemia virus type 1. Among all retroviruses analyzed, XMRV has the strongest preference for transcription start sites, CpG islands, DNase-hypersensitive sites, and gene-dense regions; all are features frequently associated with structurally open transcription regulatory regions of a chromosome. Analyses of XMRV integration sites in tissues from prostate cancer patients found a similar preference for the aforementioned chromosomal features. Additionally, XMRV integration sites in cancer tissues were associated with cancer breakpoints, common fragile sites, microRNA, and cancer-related genes, suggesting a selection process that favors certain chromosomal integration sites. In both acutely infected cells and cancer tissues, no common integration site was detected within or near proto-oncogenes or tumor suppressor genes. These results are consistent with a model in which XMRV may contribute to tumorigenicity via a paracrine mechanism.


Assuntos
Gammaretrovirus/metabolismo , Vírus da Leucemia Murina/metabolismo , Neoplasias da Próstata/virologia , Integração Viral , Animais , Linhagem Celular , Mapeamento Cromossômico , Ilhas de CpG , Endorribonucleases/genética , Endorribonucleases/metabolismo , Gammaretrovirus/genética , Humanos , Vírus da Leucemia Murina/genética , Masculino , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Dados de Sequência Molecular , Neoplasias da Próstata/genética , Sequências Reguladoras de Ácido Nucleico , Infecções por Retroviridae/metabolismo , Transcrição Gênica
18.
J Virol ; 82(19): 9789-94, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18667519

RESUMO

The env gene of gammaretroviruses encodes a glycoprotein conserved among diverse retroviruses, except for the domains involved in receptor binding. Here we show that pairs of gammaretrovirus envelope proteins (from Friend virus and GALV or xenotropic viruses) assemble into heteromers when coexpressed. This assembly results in a strong inhibition of infectivity. An unrelated envelope protein does not assemble in heteromers with the gammaretrovirus glycoproteins tested and does not affect their infectivity, demonstrating the specificity of the mechanism we describe. We propose that the numerous copies of endogenous retroviral env genes conserved within mammalian genomes act as restriction factors against infectious retroviruses.


Assuntos
Gammaretrovirus/metabolismo , Produtos do Gene env/genética , Proteínas do Envelope Viral/química , Sequência de Aminoácidos , Linhagem Celular , Genes env , Glicoproteínas/química , Humanos , Modelos Genéticos , Dados de Sequência Molecular , Receptores Virais/química , Retroviridae/genética , Homologia de Sequência de Aminoácidos , Transfecção
19.
Eur J Oral Sci ; 114(6): 517-23, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17184235

RESUMO

Different sodium-dependent inorganic phosphate (P(i)) uptake mechanisms play a major role in cellular P(i) homeostasis. The function and detailed distribution patterns of the type III Na(+)-phosphate cotransporter, PiT-2, in different organs during development are still largely unknown. We therefore examined the temporospatial expression patterns of Pit2 during murine odontogenesis. Odontoblasts were always devoid of Pit2 expression, whereas a transient, but strong, expression was detected in young secretory ameloblasts. However, the stratum intermedium and, later on, the papillary layer and cells of the subodontoblastic layer, exhibited high levels of Pit2 mRNA, which increased gradually as the tooth matured. Hormonal treatment or P(i) starvation of tooth germs in vitro did not alter Pit2 levels or patterns of expression, indicating mechanisms of regulation different from those of PiT-1 or other cell types. PiT-2 also functions as a retroviral receptor, and functional membrane-localized protein was confirmed throughout the dental papilla/pulp by demonstrating cellular permissiveness to infection by a gammaretrovirus that uses PiT-2 as a receptor. The distinct pattern of Pit2 expression during odontogenesis suggests that its P(i)-transporter function may be important for homeostasis of dental cells and not specifically for mineralization of the dental extracellular matrices. The expression of viral receptors in enamel-forming cells and the dental pulp may be of pathological significance.


Assuntos
Ameloblastos/metabolismo , Papila Dentária/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Odontogênese/fisiologia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/biossíntese , Animais , Química Encefálica , Gammaretrovirus/metabolismo , Hibridização In Situ , Camundongos , Receptores Virais/biossíntese , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/fisiologia
20.
AIDS Res Hum Retroviruses ; 22(6): 551-7, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16796530

RESUMO

Endogenous retroviruses (ERVs) probably originate from ancient germ cell infections by exogenous retroviruses. A high expression of retroviruses in reproductive tissue increases the risk of viral transmission to germ line cells. We therefore investigated the expression of human ERVs (HERVs) in normal endometrium, endometriosis, normal ovaries, and ovarian cancer. Four real-time PCRs (QPCRs) for HERV-E, HERV-I/T, HERV-H, and HERV-W, respectively, and an expression control gene were used. HERV-E RNA expression was significantly higher in endometriotic tissue (average, SD) than in normal endometrium (average, SD), both measured as ratios versus control gene expression and as. HERV-E and HERV-W RNA were higher in normal ovarian tissue than in ovarian cancer. This illustrates that HERV expression is not automatically higher in malignant tissues. The other HERV PCRs did not show expression patterns as distinctive as HERVE and HERV-W in the two kinds of reproductive tissue. A small number of candidate HERV-E loci from which the transcription took place were identified by sequencing of amplimers. The role of HERV-E and HERV-W in endometriosis merits further investigation.


Assuntos
Retrovirus Endógenos/metabolismo , Endometriose/metabolismo , Gammaretrovirus/metabolismo , Neoplasias Ovarianas/metabolismo , Sequência de Bases , Clonagem Molecular , Retrovirus Endógenos/genética , Endométrio/metabolismo , Feminino , Gammaretrovirus/genética , Humanos , Dados de Sequência Molecular , Ovário/metabolismo , Reação em Cadeia da Polimerase , RNA Viral/análise , RNA Viral/sangue , Análise de Sequência de DNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...