Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Viruses ; 14(5)2022 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-35632854

RESUMO

This review is an accompaniment to a Special Issue on "Retroviral RNA Processing". It discusses post-transcriptional regulation of retroviruses, ranging from the ancient foamy viruses to more modern viruses, such as HIV-1, HTLV-1, Rous sarcoma virus, murine leukemia virus, mouse mammary tumor virus, and Mason-Pfizer monkey virus. This review is not comprehensive. However, it tries to address some of the major questions in the field with examples of how different retroviruses express their genes. It is amazing that a single primary RNA transcript can have so many possible fates: genomic RNA, unspliced mRNA, and up to 50 different alternatively spliced mRNAs. This review will discuss the sorting of RNAs for packaging or translation, RNA nuclear export mechanisms, splicing, translation, RNA modifications, and avoidance of nonsense-mediated RNA decay.


Assuntos
RNA Viral , Retroviridae , Transporte Ativo do Núcleo Celular , Animais , Vírus da Leucemia Murina/genética , Camundongos , Processamento Pós-Transcricional do RNA , RNA Mensageiro/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Retroviridae/genética , Retroviridae/metabolismo
2.
Cell Host Microbe ; 26(2): 154-155, 2019 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-31415745

RESUMO

In this issue of Cell Host & Microbe, Courtney et al. (2019a) find that HIV-1 genomic RNA has much more m5C than cellular mRNA. Deleting the m5C "writer" NSUN2 decreases HIV-1 m5C levels, promotes translation of the HIV-1 5' gag gene, and alters splicing at the A2 site.


Assuntos
HIV-1/genética , RNA Viral , Animais , Vírus da Leucemia Murina , Metilação , Camundongos , Splicing de RNA
3.
Viruses ; 10(2)2018 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-29439385

RESUMO

Avian leukosis virus (ALV) is a simple retrovirus that can induce B-cell lymphoma in chicken(s) and other birds by insertional mutagenesis. The promoter region of telomerase reverse transcriptase (TERT) has been identified as an important integration site for tumorigenesis. Tumors with TERT promoter integrations are associated with increased TERT expression. The mechanism of this activation is still under investigation. We asked whether insertion of proviral DNA perturbs the epigenome of the integration site and, subsequently, impacts the regulation of neighboring genes. DNA cytosine methylation, which generally acts to suppress transcription, is one major form of epigenetic regulation. In this study, we examine allele-specific methylation patterns of genomic DNA from chicken tumors by bisulfite sequencing. We observed that alleles with TERT promoter integrations are associated with decreased methylation in the host genome near the site of integration. Our observations suggest that insertion of ALV in the TERT promoter region may induce expression of TERT through inhibition of maintenance methylation in the TERT promoter region.


Assuntos
Vírus da Leucose Aviária/fisiologia , Metilação de DNA , Regiões Promotoras Genéticas , Telomerase/genética , Integração Viral , Alelos , Animais , Linhagem Celular , Embrião de Galinha , Ilhas de CpG , Epigênese Genética , Provírus/genética , Sequências Repetidas Terminais , Sítio de Iniciação de Transcrição
4.
PLoS Pathog ; 13(11): e1006708, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29099869

RESUMO

Avian leukosis virus (ALV) is a simple retrovirus that causes a wide range of tumors in chickens, the most common of which are B-cell lymphomas. The viral genome integrates into the host genome and uses its strong promoter and enhancer sequences to alter the expression of nearby genes, frequently inducing tumors. In this study, we compare the preferences for ALV integration sites in cultured cells and in tumors, by analysis of over 87,000 unique integration sites. In tissue culture we observed integration was relatively random with slight preferences for genes, transcription start sites and CpG islands. We also observed a preference for integrations in or near expressed and spliced genes. The integration pattern in cultured cells changed over the course of selection for oncogenic characteristics in tumors. In comparison to tissue culture, ALV integrations are more highly selected for proximity to transcription start sites in tumors. There is also a significant selection of ALV integrations away from CpG islands in the highly clonally expanded cells in tumors. Additionally, we utilized a high throughput method to quantify the magnitude of clonality in different stages of tumorigenesis. An ALV-induced tumor carries between 700 and 3000 unique integrations, with an average of 2.3 to 4 copies of proviral DNA per infected cell. We observed increasing tumor clonality during progression of B-cell lymphomas and identified gene players (especially TERT and MYB) and biological processes involved in tumor progression.


Assuntos
Vírus da Leucose Aviária , Linfoma de Células B/virologia , Regiões Promotoras Genéticas , Integração Viral/genética , Animais , Carcinogênese , Galinhas , Células Clonais/virologia , Provírus/genética
5.
Oncotarget ; 8(34): 57302-57315, 2017 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-28915671

RESUMO

Avian leukosis virus induces tumors in chickens by integrating into the genome and altering expression of nearby genes. Thus, ALV can be used as an insertional mutagenesis tool to identify novel genes involved in tumorigenesis. Deep sequencing analysis of viral integration sites has identified CTDSPL and CTDSPL2 as common integration sites in ALV-induced B-cell lymphomas, suggesting a potential role in driving oncogenesis. We show that in tumors with integrations in these genes, the viral promoter is driving the expression of a truncated fusion transcript. Overexpression in cultured chick embryo fibroblasts reveals that CTDSPL and CTDSPL2 have oncogenic properties, including promoting cell migration. We also show that CTDSPL2 has a previously uncharacterized role in protecting cells from apoptosis induced by oxidative stress. Further, the truncated viral fusion transcripts of both CTDSPL and CTDSPL2 promote immortalization in primary cell culture.

6.
Viruses ; 9(8)2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28763028

RESUMO

All retroviruses use their full-length primary transcript as the major mRNA for Group-specific antigen (Gag) capsid proteins. This results in a long 3' untranslated region (UTR) downstream of the termination codon. In the case of Rous sarcoma virus (RSV), there is a 7 kb 3'UTR downstream of the gag terminator, containing the pol, env, and src genes. mRNAs containing long 3'UTRs, like those with premature termination codons, are frequently recognized by the cellular nonsense-mediated mRNA decay (NMD) machinery and targeted for degradation. To prevent this, RSV has evolved an RNA stability element (RSE) in the RNA immediately downstream of the gag termination codon. This 400-nt RNA sequence stabilizes premature termination codons (PTCs) in gag. It also stabilizes globin mRNAs with long 3'UTRs, when placed downstream of the termination codon. It is not clear how the RSE stabilizes the mRNA and prevents decay. We show here that the presence of RSE inhibits deadenylation severely. In addition, the RSE also impairs decapping (DCP2) and 5'-3' exonucleolytic (XRN1) function in knockdown experiments in human cells.


Assuntos
Regiões 3' não Traduzidas , Estabilidade de RNA , RNA Mensageiro/genética , RNA Viral/genética , Vírus do Sarcoma de Rous/genética , Códon de Terminação , Técnicas de Silenciamento de Genes , Produtos do Gene gag/genética , Produtos do Gene gag/metabolismo , Humanos , Degradação do RNAm Mediada por Códon sem Sentido , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Vírus do Sarcoma de Rous/metabolismo
7.
J Virol ; 91(7)2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28122976

RESUMO

All retroviruses need to integrate a DNA copy of their genome into the host chromatin. Cellular proteins regulating and targeting lentiviral and gammaretroviral integration in infected cells have been discovered, but the factors that mediate alpharetroviral avian leukosis virus (ALV) integration are unknown. In this study, we have identified the FACT protein complex, which consists of SSRP1 and Spt16, as a principal cellular binding partner of ALV integrase (IN). Biochemical experiments with purified recombinant proteins show that SSRP1 and Spt16 are able to individually bind ALV IN, but only the FACT complex effectively stimulates ALV integration activity in vitro Likewise, in infected cells, the FACT complex promotes ALV integration activity, with proviral integration frequency varying directly with cellular expression levels of the FACT complex. An increase in 2-long-terminal-repeat (2-LTR) circles in the depleted FACT complex cell line indicates that this complex regulates the ALV life cycle at the level of integration. This regulation is shown to be specific to ALV, as disruption of the FACT complex did not inhibit either lentiviral or gammaretroviral integration in infected cells.IMPORTANCE The majority of human gene therapy approaches utilize HIV-1- or murine leukemia virus (MLV)-based vectors, which preferentially integrate near genes and regulatory regions; thus, insertional mutagenesis is a substantial risk. In contrast, ALV integrates more randomly throughout the genome, which decreases the risks of deleterious integration. Understanding how ALV integration is regulated could facilitate the development of ALV-based vectors for use in human gene therapy. Here we show that the FACT complex directly binds and regulates ALV integration efficiency in vitro and in infected cells.


Assuntos
Vírus da Leucose Aviária/genética , Proteínas de Ciclo Celular/fisiologia , DNA Viral/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proteínas de Grupo de Alta Mobilidade/fisiologia , Fatores de Transcrição/fisiologia , Fatores de Elongação da Transcrição/fisiologia , Sequência de Aminoácidos , Animais , Vírus da Leucose Aviária/enzimologia , Embrião de Galinha , Sequência Conservada , Células HEK293 , Humanos , Integrases/fisiologia , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Integração Viral
8.
Noncoding RNA ; 4(1)2017 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-29657298

RESUMO

A novel antisense transcript was identified in the human telomerase reverse transcriptase (hTERT) promoter region, suggesting that the hTERT promoter is bidirectional. This transcript, named hTERT antisense promoter-associated (hTAPAS) RNA, is a 1.6 kb long non-coding RNA. hTAPAS transcription is initiated 167 nucleotides upstream of the hTERT transcription start site and is present in both the nucleus and the cytoplasm. Surprisingly, we observed that a large fraction of the hTERT polyadenylated RNA is localized in the nucleus, suggesting this might be an additional means of regulating the cellular abundance of hTERT protein. Both hTAPAS and hTERT are expressed in immortalized B-cells and human embryonic stem cells but are not detected in normal somatic cells. hTAPAS expression inversely correlates with hTERT expression in different types of cancer samples. Moreover, hTAPAS expression is not promoted by an hTERT promoter mutation (-124 C>T). Antisense-oligonucleotide mediated knockdown of hTAPAS results in an increase in hTERT expression. Conversely, ectopic overexpression of hTAPAS down regulates hTERT expression, suggesting a negative role in hTERT gene regulation. These observations provide insights into hTAPAS as a novel player that negatively regulates hTERT expression and may be involved in telomere length homeostasis.

9.
Genes (Basel) ; 7(11)2016 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-27792139

RESUMO

Non-Hodgkin lymphomas (NHL) are a heterogeneous group of immune cell neoplasms that comprise molecularly distinct lymphoma subtypes. Recent work has identified high frequency promoter point mutations in the telomerase reverse transcriptase (TERT) gene of different cancer types, including melanoma, glioma, liver and bladder cancer. TERT promoter mutations appear to correlate with increased TERT expression and telomerase activity in these cancers. In contrast, breast, pancreatic, and prostate cancer rarely demonstrate mutations in this region of the gene. TERT promoter mutation prevalence in NHL has not been thoroughly tested thus far. We screened 105 B-cell lymphoid malignancies encompassing nine NHL subtypes and acute lymphoblastic leukemia, for TERT promoter mutations. Our results suggest that TERT promoter mutations are rare or absent in most NHL. Thus, the classical TERT promoter mutations may not play a major oncogenic role in TERT expression and telomerase activation in NHL.

10.
J Virol ; 90(20): 9509-17, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27512065

RESUMO

UNLABELLED: Avian leukosis virus (ALV) induces tumors by integrating its proviral DNA into the chicken genome and altering the expression of nearby genes via strong promoter and enhancer elements. Viral integration sites that contribute to oncogenesis are selected in tumor cells. Deep-sequencing analysis of B-cell lymphoma DNA confirmed that the telomerase reverse transcriptase (TERT) gene promoter is a common ALV integration target. Twenty-six unique proviral integration sites were mapped between 46 and 3,552 nucleotides (nt) upstream of the TERT transcription start site, predominantly in the opposite transcriptional orientation to TERT Transcriptome-sequencing (RNA-seq) analysis of normal bursa revealed a transcribed region upstream of TERT in the opposite orientation, suggesting the TERT promoter is bidirectional. This transcript appears to be an uncharacterized antisense RNA. We have previously shown that TERT expression is upregulated in tumors with integrations in the TERT promoter region. We now report that the viral promoter drives the expression of a chimeric transcript containing viral sequences spliced to exons 4 through 7 of this antisense RNA. Clonal expansion of cells with ALV integrations driving overexpression of the TERT antisense RNA suggest it may have a role in tumorigenesis. IMPORTANCE: The data suggest that ALV integrations in the TERT promoter region drive the overexpression of a novel antisense RNA and contribute to the development of lymphomas.


Assuntos
Vírus da Leucose Aviária/genética , Leucose Aviária/genética , Leucose Aviária/virologia , Linfoma de Células B/genética , Linfoma de Células B/virologia , RNA Antissenso/genética , Telomerase/genética , Animais , Transformação Celular Neoplásica/genética , Galinhas , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Regiões Promotoras Genéticas/genética , Sítio de Iniciação de Transcrição/fisiologia , Transcriptoma/genética , Regulação para Cima/genética , Integração Viral/genética
11.
Elife ; 52016 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-26744779

RESUMO

The nonsense-mediated mRNA decay (NMD) pathway degrades mRNAs containing long 3'UTRs to perform dual roles in mRNA quality control and gene expression regulation. However, expansion of vertebrate 3'UTR functions has required a physical expansion of 3'UTR lengths, complicating the process of detecting nonsense mutations. We show that the polypyrimidine tract binding protein 1 (PTBP1) shields specific retroviral and cellular transcripts from NMD. When bound near a stop codon, PTBP1 blocks the NMD protein UPF1 from binding 3'UTRs. PTBP1 can thus mark specific stop codons as genuine, preserving both the ability of NMD to accurately detect aberrant mRNAs and the capacity of long 3'UTRs to regulate gene expression. Illustrating the wide scope of this mechanism, we use RNA-seq and transcriptome-wide analysis of PTBP1 binding sites to show that many human mRNAs are protected by PTBP1 and that PTBP1 enrichment near stop codons correlates with 3'UTR length and resistance to NMD.


Assuntos
Regiões 3' não Traduzidas , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Degradação do RNAm Mediada por Códon sem Sentido , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , RNA Mensageiro/metabolismo , Sítios de Ligação , Códon sem Sentido , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , RNA Helicases , Análise de Sequência de RNA , Transativadores/antagonistas & inibidores
12.
mBio ; 6(6): e01863-15, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26670384

RESUMO

UNLABELLED: Avian leukosis virus (ALV) induces B-cell lymphoma and other neoplasms in chickens by integrating within or near cancer genes and perturbing their expression. Four genes--MYC, MYB, Mir-155, and TERT--have previously been identified as common integration sites in these virus-induced lymphomas and are thought to play a causal role in tumorigenesis. In this study, we employ high-throughput sequencing to identify additional genes driving tumorigenesis in ALV-induced B-cell lymphomas. In addition to the four genes implicated previously, we identify other genes as common integration sites, including TNFRSF1A, MEF2C, CTDSPL, TAB2, RUNX1, MLL5, CXorf57, and BACH2. We also analyze the genome-wide ALV integration landscape in vivo and find increased frequency of ALV integration near transcriptional start sites and within transcripts. Previous work has shown ALV prefers a weak consensus sequence for integration in cultured human cells. We confirm this consensus sequence for ALV integration in vivo in the chicken genome. IMPORTANCE: Avian leukosis virus induces B-cell lymphomas in chickens. Earlier studies showed that ALV can induce tumors through insertional mutagenesis, and several genes have been implicated in the development of these tumors. In this study, we use high-throughput sequencing to reveal the genome-wide ALV integration landscape in ALV-induced B-cell lymphomas. We find elevated levels of ALV integration near transcription start sites and use common integration site analysis to greatly expand the number of genes implicated in the development of these tumors. Interestingly, we identify several genes targeted by viral insertions that have not been previously shown to be involved in cancer.


Assuntos
Vírus da Leucose Aviária/fisiologia , Linfoma de Células B/virologia , Integração Viral , Animais , Galinhas , Sequenciamento de Nucleotídeos em Larga Escala , Sítio de Iniciação de Transcrição , Transcrição Gênica
13.
Genome Announc ; 3(2)2015 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-25858851

RESUMO

We report the complete genome sequence of avian leukosis virus subgroup J (ALV-J) isolate PDRC-59831, which causes myeloid leukosis and hemangiomas in chickens. This is an American ALV-J isolate, which was found in a 38-week-old broiler breeder chicken on a farm in Georgia in 2007.

14.
RNA ; 21(4): 564, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25780139
15.
J Virol ; 89(9): 4712-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25673726

RESUMO

UNLABELLED: Avian leukosis virus subgroup J (ALV-J) is a simple retrovirus that can cause hemangiomas and myeloid tumors in chickens and is currently a major economic problem in Asia. Here we characterize ALV-J strain PDRC-59831, a newly studied U.S. isolate of ALV-J. Five-day-old chicken embryos were infected with this virus, and the chickens developed myeloid leukosis and hemangiomas within 2 months after hatching. To investigate the mechanism of pathogenesis, we employed high-throughput sequencing to analyze proviral integration sites in these tumors. We found expanded clones with integrations in the MET gene in two of the five hemangiomas studied. This integration locus was not seen in previous work characterizing ALV-J-induced myeloid leukosis. MET is a known proto-oncogene that acts through a diverse set of signaling pathways and is involved in many neoplasms. We show that tumors harboring MET integrations exhibit strong overexpression of MET mRNA. IMPORTANCE: These data suggest that ALV-J induces oncogenesis by insertional mutagenesis, and integrations in the MET oncogene can drive the overexpression of MET and contribute to the development of hemangiomas.


Assuntos
Vírus da Leucose Aviária/fisiologia , Leucose Aviária/virologia , Hemangioma/virologia , Proteínas Proto-Oncogênicas c-met/genética , Integração Viral , Animais , Vírus da Leucose Aviária/isolamento & purificação , Galinhas , Sequenciamento de Nucleotídeos em Larga Escala , Estados Unidos
16.
Curr Opin Microbiol ; 18: 78-82, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24632073

RESUMO

Unspliced Rous sarcoma virus (RSV) retroviral mRNA undergoes nonsense-mediated RNA decay (NMD) if it has premature termination codons in the gag gene. However, its normal gag termination codon is not subject to NMD despite being 7kb from the 3' poly(A) sequence. An RNA stability element (RSE) has been identified immediately downstream of gag in the RSV genome. It appears to determine the proper context for translation termination and protects the RNA from NMD. The viral stability element may prevent Up-frameshift 1 (Upf1) protein from interacting with the terminating ribosome and release factors to initiate NMD.


Assuntos
Degradação do RNAm Mediada por Códon sem Sentido , RNA Viral/genética , RNA Viral/metabolismo , Vírus do Sarcoma de Rous/genética , Códon sem Sentido , Produtos do Gene gag/genética
17.
Curr Opin Virol ; 3(6): 664-9, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24011707

RESUMO

Avian retroviruses were originally identified as cancer-inducting filterable agents in chicken neoplasms at the beginning of the 20th century. Since their discovery, the study of these simple retroviruses has contributed greatly to our understanding of viral replication and cancer. Avian retroviruses continue to evolve and have great economic importance in the poultry industry worldwide. The aim of this review is to provide a broad overview of the genome, pathology, and replication of avian retroviruses. Notable gaps in our current knowledge are highlighted, and areas where avian retroviruses differ from other retroviruses are emphasized.


Assuntos
Retroviridae/fisiologia , Replicação Viral , Animais , Transformação Celular Viral , Genoma Viral , Interações Hospedeiro-Patógeno , Aves Domésticas , Retroviridae/genética
18.
Wiley Interdiscip Rev RNA ; 4(5): 567-80, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23754689

RESUMO

After reverse transcription of the retroviral RNA genome and integration of the DNA provirus into the host genome, host machinery is used for viral gene expression along with viral proteins and RNA regulatory elements. Here, we discuss co-transcriptional and posttranscriptional regulation of retroviral gene expression, comparing simple and complex retroviruses. Cellular RNA polymerase II synthesizes full-length viral primary RNA transcripts that are capped and polyadenylated. All retroviruses generate a singly spliced env mRNA from this primary transcript, which encodes the viral glycoproteins. In addition, complex viral RNAs are alternatively spliced to generate accessory proteins, such as Rev, which is involved in posttranscriptional regulation of HIV-1 RNA. Importantly, the splicing of all retroviruses is incomplete; they must maintain and export a fraction of their primary RNA transcripts. This unspliced RNA functions both as the major mRNA for Gag and Pol proteins and as the packaged genomic RNA. Different retroviruses export their unspliced viral RNA from the nucleus to the cytoplasm by either Tap-dependent or Rev/CRM1-dependent routes. Translation of the unspliced mRNA involves frame-shifting or termination codon suppression so that the Gag proteins, which make up the capsid, are expressed more abundantly than the Pol proteins, which are the viral enzymes. After the viral polyproteins assemble into viral particles and bud from the cell membrane, a viral encoded protease cleaves them. Some retroviruses have evolved mechanisms to protect their unspliced RNA from decay by nonsense-mediated RNA decay and to prevent genome editing by the cellular APOBEC deaminases.


Assuntos
Regulação Viral da Expressão Gênica , Precursores de RNA/metabolismo , RNA Viral/metabolismo , Retroviridae/genética , Biossíntese de Proteínas , Processamento Pós-Transcricional do RNA , Transcrição Gênica
20.
PLoS One ; 7(10): e48016, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23110161

RESUMO

Telomeric sequences are added by an enzyme called telomerase that is made of two components: a catalytic protein called telomerase reverse transcriptase (TERT) and an integral RNA template (TR). Telomerase expression is tightly regulated at each step of gene expression, including alternative splicing of TERT mRNA. While over a dozen different alternative splicing events have been reported for human TERT mRNA, these were all in the 3' half of the coding region. We were interested in examining splicing of the 5' half of hTERT mRNA, especially since exon 2 is unusually large (1.3 kb). Internal mammalian exons are usually short, typically only 50 to 300 nucleotides, and most long internal exons are alternatively processed. We used quantitative RT-PCR and high-throughput sequencing data to examine the variety and quantity of mRNA species generated from the hTERT locus. We determined that there are approximately 20-40 molecules of hTERT mRNA per cell in the A431 human cell line. In addition, we describe an abundant, alternatively-spliced mRNA variant that excludes TERT exon 2 and was seen in other primates. This variant causes a frameshift and results in translation termination in exon 3, generating a 12 kDa polypeptide.


Assuntos
Éxons/genética , Splicing de RNA , RNA Mensageiro/genética , Telomerase/genética , Animais , Linfócitos B/enzimologia , Linfócitos B/metabolismo , Sequência de Bases , Células COS , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Camundongos , Células NIH 3T3 , Filogenia , Primatas , RNA/genética , RNA/metabolismo , RNA Mensageiro/classificação , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência do Ácido Nucleico , Telomerase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...