Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34103391

RESUMO

As COVID-19 continues to spread across the world, it is increasingly important to understand the factors that influence its transmission. Seasonal variation driven by responses to changing environment has been shown to affect the transmission intensity of several coronaviruses. However, the impact of the environment on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remains largely unknown, and thus seasonal variation remains a source of uncertainty in forecasts of SARS-CoV-2 transmission. Here we address this issue by assessing the association of temperature, humidity, ultraviolet radiation, and population density with estimates of transmission rate (R). Using data from the United States, we explore correlates of transmission across US states using comparative regression and integrative epidemiological modeling. We find that policy intervention ("lockdown") and reductions in individuals' mobility are the major predictors of SARS-CoV-2 transmission rates, but, in their absence, lower temperatures and higher population densities are correlated with increased SARS-CoV-2 transmission. Our results show that summer weather cannot be considered a substitute for mitigation policies, but that lower autumn and winter temperatures may lead to an increase in transmission intensity in the absence of policy interventions or behavioral changes. We outline how this information may improve the forecasting of COVID-19, reveal its future seasonal dynamics, and inform intervention policies.


Assuntos
COVID-19/transmissão , Temperatura Baixa , Densidade Demográfica , Número Básico de Reprodução , COVID-19/epidemiologia , COVID-19/prevenção & controle , Controle de Doenças Transmissíveis/legislação & jurisprudência , Previsões , Humanos , Movimento , SARS-CoV-2 , Estações do Ano , Estados Unidos/epidemiologia
2.
J Gen Virol ; 102(12)2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34939563

RESUMO

Viruses in the family Retroviridae are found in a wide variety of vertebrate hosts. Enveloped virions are 80-100 nm in diameter with an inner core containing the viral genome and replicative enzymes. Core morphology is often characteristic for viruses within the same genus. Replication involves reverse transcription and integration into host cell DNA, resulting in a provirus. Integration into germline cells can result in a heritable provirus known as an endogenous retrovirus. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the family Retroviridae, which is available at ictv.global/report/retroviridae.


Assuntos
Vírus de DNA/classificação , Retroviridae/classificação , Animais , Vírus de DNA/genética , Vírus de DNA/fisiologia , Vírus de DNA/ultraestrutura , Genoma Viral , Especificidade de Hospedeiro , Retroviridae/genética , Retroviridae/fisiologia , Retroviridae/ultraestrutura , Vertebrados/virologia , Vírion/ultraestrutura , Replicação Viral
3.
Retrovirology ; 15(1): 59, 2018 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-30153831

RESUMO

Retroviral integration into germline DNA can result in the formation of a vertically inherited proviral sequence called an endogenous retrovirus (ERV). Over the course of their evolution, vertebrate genomes have accumulated many thousands of ERV loci. These sequences provide useful retrospective information about ancient retroviruses, and have also played an important role in shaping the evolution of vertebrate genomes. There is an immediate need for a unified system of nomenclature for ERV loci, not only to assist genome annotation, but also to facilitate research on ERVs and their impact on genome biology and evolution. In this review, we examine how ERV nomenclatures have developed, and consider the possibilities for the implementation of a systematic approach for naming ERV loci. We propose that such a nomenclature should not only provide unique identifiers for individual loci, but also denote orthologous relationships between ERVs in different species. In addition, we propose that-where possible-mnemonic links to previous, well-established names for ERV loci and groups should be retained. We show how this approach can be applied and integrated into existing taxonomic and nomenclature schemes for retroviruses, ERVs and transposable elements.


Assuntos
Retrovirus Endógenos/classificação , Retrovirus Endógenos/genética , Animais , Evolução Molecular , Loci Gênicos , Variação Genética , Genômica , Humanos , Terminologia como Assunto , Vertebrados/genética , Vertebrados/virologia
4.
J Virol ; 88(21): 12907-9, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25142605

RESUMO

Following the recent availability of high-coverage genomes for Denisovan and Neanderthal hominids, we conducted a screen for endogenized retroviruses, identifying six novel, previously unreported HERV-K(HML2) elements (HERV-K is human endogenous retrovirus K). These elements are absent from the human genome (hg38) and appear to be unique to archaic hominids. These findings provide further evidence supporting the recent activity of the HERV-K(HML2) group, which has been implicated in human disease. They will also provide insights into the evolution of archaic hominids.


Assuntos
Retrovirus Endógenos/genética , Retrovirus Endógenos/isolamento & purificação , Fósseis/virologia , Hominidae/virologia , Animais , Retrovirus Endógenos/classificação , Feminino , Genoma
6.
Sci Rep ; 14(1): 13326, 2024 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-38858479

RESUMO

Previous work has shown that environmental variables affect SARS-CoV-2 transmission, but it is unclear whether different strains show similar environmental responses. Here we leverage genetic data on the transmission of three (Alpha, Delta and Omicron BA.1) variants of SARS-CoV-2 throughout England, to unpick the roles that climate and public-health interventions play in the circulation of this virus. We find evidence for enhanced transmission of the virus in colder conditions in the first variant selective sweep (of Alpha, in winter), but limited evidence of an impact of climate in either the second (of Delta, in the summer, when vaccines were prevalent) or third sweep (of Omicron, in the winter, during a successful booster-vaccination campaign). We argue that the results for Alpha are to be expected if the impact of climate is non-linear: we find evidence of an asymptotic impact of temperature on the alpha variant transmission rate. That is, at lower temperatures, the influence of temperature on transmission is much higher than at warmer temperatures. As with the initial spread of SARS-CoV-2, however, the overwhelming majority of variation in disease transmission is explained by the intrinsic biology of the virus and public-health mitigation measures. Specifically, when vaccination rates are high, a major driver of the spread of a new variant is it's ability to evade immunity, and any climate effects are secondary (as evidenced for Delta and Omicron). Climate alone cannot describe the transmission dynamics of emerging SARS-CoV-2 variants.


Assuntos
COVID-19 , SARS-CoV-2 , SARS-CoV-2/genética , SARS-CoV-2/isolamento & purificação , SARS-CoV-2/imunologia , COVID-19/transmissão , COVID-19/virologia , COVID-19/epidemiologia , COVID-19/prevenção & controle , Humanos , Inglaterra/epidemiologia , Estações do Ano , Temperatura , Clima , Vacinas contra COVID-19/imunologia
7.
BMC Evol Biol ; 11: 139, 2011 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-21609472

RESUMO

BACKGROUND: Porcine endogenous retroviruses (PERVs) represent remnants of an exogenous form that have become integrated in the domestic pig (Sus scrofa) genome. Although they are usually inactive, the capacity of γ1 ERVs to infect human cells in vitro has raised concerns about xenotransplantation because the viruses could cross the species barrier to humans. Here we have analyzed the evolution of γ1 ERVs in ten species of Suidae (suids, pigs and hogs) from Eurasia and Africa using DNA sequences for their coding domains (gag, pro/pol and env genes). For comparison with γ1 PERVs, we have also analysed γ2 ERVs which in domestic pigs are known to be inactive and do not pose a risk to xenotransplantation. RESULTS: Phylogenetic analysis using Bayesian inference showed that γ1 and γ2 ERVs have distinctive evolutionary histories. Firstly, two different viral lineages of γ1 ERVs were found and a coevolutionary analysis demonstrated that they correspond broadly to their host phylogeny, one of Eurasian and another of African species, and show no evidence of horizontal transmission. γ2 ERVs, however, show a bush-like evolution, suggesting a rapid viral radiation from a single common ancestor with no correspondence between host and viral evolutionary trees. Furthermore, though γ1 ERV env genes do not possess frequent stop codons, γ2 env genes do. To understand whether γ1 suid ERVs may be still replicating, we have also evaluated their likely mechanism of proliferation by statistically testing internal to terminal branches using nonsynonymous versus synonymous substitution ratios. Our results suggest that γ1 ERVs are increasing in copy number by reinfection, which requires the translocation of the virus from one cell to another. CONCLUSIONS: Evidence of at least two viral subpopulations was observed in γ1 ERVs from Eurasian and African host species. These results should be taken into account in xenotransplantation since γ1 ERVs appear to be codiverging with their host and maintaining ongoing capacity to infect somatic and germ cells.


Assuntos
Gammaretrovirus/genética , Gammaretrovirus/fisiologia , Interações Hospedeiro-Patógeno , Suínos/virologia , África , Animais , Ásia , Teorema de Bayes , Europa (Continente) , Genes env , Genes gag , Genes pol , Dados de Sequência Molecular , Filogenia , Recombinação Genética
8.
Proc Natl Acad Sci U S A ; 105(51): 20362-7, 2008 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-19075221

RESUMO

Lentiviruses chronically infect a broad range of mammalian species and have been transmitted from primates to humans, giving rise to multiple outbreaks of HIV infection over the past century. Although the circumstances surrounding these recent zoonoses are becoming clearer, the nature and timescale of interaction between lentiviruses and primates remains unknown. Here, we report the discovery of an endogenous lentivirus in the genome of the gray mouse lemur (Microcebus murinus), a strepsirrhine primate from Madagascar, demonstrating that lentiviruses are capable of invading the primate germ line. Phylogenetic analysis places gray mouse lemur prosimian immunodeficiency virus (pSIVgml) basal to all known primate lentiviruses and, consistent with this, its genomic organization is intermediate between the nonprimate lentiviruses and their more derived primate counterparts. Thus, pSIVgml represents the first unambiguous example of a viral transitional form, revealing the acquisition and loss of genomic features during lentiviral evolution. Furthermore, because terrestrial mammal populations in Madagascar and Africa are likely to have been isolated from one another for at least 14 million years, the presence of pSIVgml in the gray mouse lemur genome indicates that lentiviruses must have been infecting primates for at least this period of time, or have been transmitted between Malagasy and African primate populations by a vector species capable of traversing the Mozambique channel. The discovery of pSIVgml illustrates the utility of endogenous sequences for the study of contemporary retroviruses and indicates that primate lentiviruses may be considerably older and more broadly distributed than previously thought.


Assuntos
Evolução Biológica , Lemur/virologia , Lentivirus de Primatas/genética , Provírus/genética , África , Animais , Sequência de Bases , Genoma/genética , Lemur/genética , Madagáscar , Dados de Sequência Molecular , Filogenia
9.
Curr Biol ; 13(17): 1518-23, 2003 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-12956954

RESUMO

One subset of sequences present within mammalian genomes is the retroelements, which include endogenous retroviruses and retrotransposons. While there are typically thousands of copies of endogenous retroviruses within mammalian hosts, almost no LTR-retrotransposon-like sequences have been identified. Here, we report the presence of a remarkably intact and conserved gypsy-type LTR-retrotransposon sequence within the genomes of several mammals, including humans and mice. Each host probably contains a single orthologous element, indicating that the original, ancestral gypsy LTR-retrotransposon first integrated into mammals over 70 million years ago. It is thus the first described example of a near-intact orthologous retroelement within humans and mice and is one of the most ancient retroelement sequences described to date. Despite their extreme age, the orthologs within each species examined contain a large ORF, between 4.0 and 5.2 kb in length, encoding proteins with sequence similarity to LTR-retrotransposon-derived Capsid (CA), Protease (PR), Reverse Transcriptase (RT), RibonucleaseH (RNaseH), and Integrase (IN). Calculation of nonsynonymous and synonymous nucleotide substitution frequencies indicated that the encoded proteins are under purifying selection, suggesting that these elements have, in fact, been co-opted by their hosts. A possible function for these elements, involving gypsy LTR-retrotransposon restriction in mammals, is discussed.


Assuntos
Evolução Molecular , Mamíferos/genética , Filogenia , Retroelementos/genética , Sequências Repetidas Terminais/genética , Sequência de Aminoácidos , Animais , Mapeamento Cromossômico , Genoma , Humanos , Camundongos/genética , Dados de Sequência Molecular , Ratos/genética , Seleção Genética , Alinhamento de Sequência , Homologia de Sequência
10.
Philos Trans R Soc Lond B Biol Sci ; 368(1626): 20120503, 2013 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-23938752

RESUMO

The evolutionary arms race between mammals and retroviruses has long been recognized as one of the oldest host-parasite interactions. Rapid evolution rates in exogenous retroviruses have often made accurate viral age estimations highly problematic. Endogenous retroviruses (ERVs), however, integrate into the germline of their hosts, and are subjected to their evolutionary rates. This study describes, for the first time, a retroviral orthologue predating the divergence of placental mammals, giving it a minimum age of 104-110 Myr. Simultaneously, other orthologous selfish genetic elements (SGEs), inserted into the ERV sequence, provide evidence for the oldest individual mammalian-wide interspersed repeat and medium-reiteration frequency interspersed repeat mammalian repeats, with the same minimum age. The combined use of shared SGEs and reconstruction of viral orthologies defines new limits and increases maximum 'lookback' times, with subsequent implications for the field of paleovirology.


Assuntos
Retrovirus Endógenos/genética , Genoma , Interações Hospedeiro-Patógeno/genética , Mamíferos/genética , Mamíferos/virologia , Animais , Sequência de Bases , Dados de Sequência Molecular , Filogenia , Alinhamento de Sequência
11.
Infect Genet Evol ; 11(3): 686-93, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21256982

RESUMO

Diversity of long terminal repeats (LTRs) from γ1 endogenous retroviruses (ERVs) was analysed by DNA sequencing in 10 species of the family Suidae (suids, pigs and hogs). Phylogenetic analysis separated LTR sequences into two groups which correlated approximately with either the previously described cluster I and III, or the clusters II, IV and V. Interestingly, a specific LTR exhibiting a novel molecular rearrangement was identified exclusively within African host species when compared to LTRs previously reported from known ERVs in the domestic pig (Sus scrofa). Furthermore, other sections of LTRs appear to be unique to African suids as suggested by phylogenetic analysis. These differences between African and Eurasian ERV lineages show that these ERVs belong to different viral sub-populations, implying coevolution of endogenous viral sequences with their host species and providing no evidence of transfer of viral sequences between African and Eurasian suids.


Assuntos
Retrovirus Endógenos/genética , Variação Genética , Suínos/virologia , Sequências Repetidas Terminais , África , Animais , Ásia , Retrovirus Endógenos/classificação , Europa (Continente) , Funções Verossimilhança , Modelos Genéticos , Dados de Sequência Molecular , Filogenia , Alinhamento de Sequência , Análise de Sequência de DNA , Proteínas do Envelope Viral/genética
12.
Science ; 325(5947): 1512, 2009 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-19762636

RESUMO

Retroviruses can leave a "fossil record" in their hosts' genomes in the form of endogenous retroviruses. Foamy viruses, complex retroviruses that infect mammals, have been notably absent from this record. We have found an endogenous foamy virus within the genomes of sloths and show that foamy viruses were infecting mammals more than 100 million years ago and codiverged with their hosts across an entire geological era. Our analysis highlights the role of evolutionary constraint in maintaining viral genome structure and indicates that accessory genes and mammalian mechanisms of innate immunity are the products of macroevolutionary conflict played out over a geological time scale.


Assuntos
Evolução Biológica , Retrovirus Endógenos/genética , Evolução Molecular , Bichos-Preguiça/genética , Bichos-Preguiça/virologia , Spumavirus/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Retrovirus Endógenos/classificação , Genoma , Genoma Viral , Imunidade Inata , Dados de Sequência Molecular , Filogenia , Infecções por Retroviridae/veterinária , Infecções por Retroviridae/virologia , Bichos-Preguiça/classificação , Bichos-Preguiça/imunologia , Spumavirus/classificação , Tempo
13.
J Virol ; 81(19): 10712-7, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17634225

RESUMO

Endogenous retroviruses (ERVs) result from germ line infections by exogenous retroviruses. They can proliferate within the genome of their host species until they are either inactivated by mutation or removed by recombinational deletion. ERVs belong to a diverse group of mobile genetic elements collectively termed transposable elements (TEs). Numerous studies have attempted to elucidate the factors determining the genomic distribution and persistence of TEs. Here we show that, within humans, gene density and not recombination rate correlates with fixation of endogenous retroviruses, whereas the local recombination rate determines their persistence in a full-length state. Recombination does not appear to influence fixation either via the ectopic exchange model or by indirect models based on the efficacy of selection. We propose a model linking rates of meiotic recombination to the probability of recombinational deletion to explain the effect of recombination rate on persistence. Chromosomes 19 and Y are exceptions, possessing more elements than other regions, and we suggest this is due to low gene density and elevated rates of human ERV integration in males for chromosome Y and segmental duplication for chromosome 19.


Assuntos
Retrovirus Endógenos/genética , Genoma Humano/genética , Recombinação Genética , Cromossomos Humanos Par 19/genética , Cromossomos Humanos Y/genética , Humanos
14.
Proc Natl Acad Sci U S A ; 104(15): 6261-5, 2007 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-17384150

RESUMO

The lentiviruses are associated with a wide range of chronic diseases in mammals. These include immunodeficiencies (such as HIV/AIDS in humans), malignancies, and lymphatic and neurological disorders in primates, felids, and a variety of wild and domesticated ungulates. Evolutionary analyses of the genomic sequences of modern-day lentiviruses have suggested a relatively recent date for their emergence, but the failure to identify any endogenous, vertically transmitted examples has meant that their longer term evolutionary history and origin remain unknown. Here we report the discovery and characterization of retroviral sequences belonging to a new lentiviral subgroup from the European rabbit (Oryctolagus cuniculus). These viruses, the first endogenous examples described, are >7 million years old and thus provide the first evidence for an ancient origin of the lentiviruses. Despite being ancient, this subgroup contains many of the features found in present-day lentiviruses, such as the presence of tat and rev genes, thus also indicating an ancient origin for the complex regulation of lentivirus gene expression. Although the virus we describe is defective, reconstruction of an infectious progenitor could provide novel insights into lentivirus biology and host interactions.


Assuntos
Retrovirus Endógenos/genética , Evolução Molecular , Genoma/genética , Lentivirus/genética , Coelhos/virologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Biologia Computacional , Genes Virais/genética , Componentes Genômicos , Genômica/métodos , Humanos , Lentivirus/classificação , Funções Verossimilhança , Modelos Genéticos , Dados de Sequência Molecular , Análise de Sequência de DNA
15.
J Virol ; 81(17): 9437-42, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17581995

RESUMO

The fate of most human endogenous retroviruses (HERVs) has been to undergo recombinational deletion. This process involves homologous recombination between the flanking long terminal repeats (LTRs) of a full-length element, leaving a relic structure in the genome termed a solo LTR. We examined loci in one family, HERV-K(HML2), and found that the deletion rate decreased markedly with age: the rate among recently integrated loci was almost 200-fold higher than that among loci whose insertion predated the divergence of humans and chimpanzees (8 x 10(-5) and 4 x 10(-7) recombinational deletion events per locus per generation, respectively). One hypothesis for this finding is that increasing mutational divergence between the flanking LTRs reduces the probability of homologous recombination and thus the rate of solo LTR formation. Consistent with this idea, we were able to replicate the observed rates by a simulation in which the probability of recombinational deletion was reduced 10-fold by a single mutation and 100-fold by any additional mutations. We also discuss the evidence for other factors that may influence the relationship between locus age and the rate of deletion, for example, host recombination rates and selection, and highlight the consequences of recombinational deletion for dating recent HERV integrations.


Assuntos
DNA Viral/genética , Retrovirus Endógenos/genética , Recombinação Genética , Deleção de Sequência , Biologia Computacional , Humanos , Sequências Repetidas Terminais , Fatores de Tempo
16.
J Virol ; 79(19): 12507-14, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16160178

RESUMO

The published human genome sequence contains many thousands of endogenous retroviruses (HERVs) but all are defective, containing nonsense mutations or major deletions. Only the HERV-K(HML2) family has been active since the divergence of humans and chimpanzees; it contains many members that are human specific, as well as several that are insertionally polymorphic (an inserted element present only in some human individuals). Here we perform a genomewide survey of insertional polymorphism levels in this family by using the published human genome sequence and a diverse sample of 19 humans. We find that there are 113 human-specific HERV-K(HML2) elements in the human genome sequence, 8 of which are insertionally polymorphic (11 if we extrapolate to those within regions of the genome that were not suitable for amplification). The average rate of accumulation since the divergence with chimpanzees is thus approximately 3.8 x 10(-4) per haploid genome per generation. Furthermore, we find that the number of polymorphic elements is not significantly different from that predicted by a standard population genetic model that assumes constant activity of the family until the present. This suggests to us that the HERV-K(HML2) family may be active in present-day humans. Active (replication-competent) elements are likely to have inserted very recently and to be present at low allele frequencies, and they may be causing disease in the individuals carrying them. This view of the family from a population perspective rather than a genome perspective will inform the current debate about a possible role of HERV-K(HML2) in human disease.


Assuntos
DNA Viral/genética , Retrovirus Endógenos/genética , Genoma Humano , Polimorfismo Genético , Integração Viral , Evolução Molecular , Feminino , Genoma Viral , Humanos , Masculino , Homologia de Sequência , Sequências Repetidas Terminais
17.
Mol Biol Evol ; 22(4): 814-7, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15659556

RESUMO

There are at least 31 families of human endogenous retroviruses (HERVs), each derived from an independent infection by an exogenous virus. Using evidence of purifying selection on HERV genes, we have shown previously that reinfection by replication-competent elements was the predominant mechanism of copying in some families. Here we analyze the evolution of 17 HERV families using d(N)/d(S) ratios and find a positive relationship between copy number and the use of additional copying mechanisms. All families with more than 200 elements have also used one or more of the following mechanisms: (1) complementation in trans (elements copied by other elements of the same family; HERV-H and ERV-9), (2) retrotransposition in cis (elements copying themselves) within germ-line cells (HERV-K(HML3)), and (3) being copied by non-HERV machinery (HERV-W). We discuss why these other mechanisms are rare in most families and suggest why complementation in trans is significant only in the larger families.


Assuntos
Retrovirus Endógenos/genética , Infecções por Retroviridae/genética , Retrovirus Endógenos/classificação , Funções Verossimilhança , Filogenia , Sequências Repetitivas de Ácido Nucleico , Integração Viral
18.
J Virol ; 79(10): 6478-86, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15858031

RESUMO

Endogenous retroviruses (ERVs) are widespread in vertebrate genomes and have been loosely grouped into "classes" on the basis of their phylogenetic relatedness to the established genera of exogenous retroviruses. Four of these genera-the lentiviruses, alpharetroviruses, betaretroviruses, and deltaretroviruses-form a well-supported clade in retroviral phylogenies, and ERVs that group with these genera have been termed class II ERVs. We used PCR amplification and sequencing of retroviral fragments from more than 130 vertebrate taxa to investigate the evolution of the class II retroviruses in detail. We confirm that class II retroviruses are largely confined to mammalian and avian hosts and provide evidence for a major novel group of avian retroviruses, and we identify additional members of both the alpha- and the betaretrovirus genera. Phylogenetic analyses demonstrated that the avian and mammalian viruses form distinct monophyletic groups, implying that interclass transmission has occurred only rarely during the evolution of the class II retroviruses. In contrast to previous reports, the lentiviruses clustered as sister taxa to several endogenous retroviruses derived from rodents and insectivores. This topology was further supported by the shared loss of both the class II PR-Pol frameshift site and the class II retrovirus G-patch domain.


Assuntos
Retrovirus Endógenos/classificação , Genoma Viral , Animais , Evolução Biológica , Retrovirus Endógenos/genética , Lentivirus/genética , Dados de Sequência Molecular , Vertebrados/virologia
19.
Virus Genes ; 26(3): 291-315, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12876457

RESUMO

The retroviral capacity for integration into the host genome can give rise to endogenous retroviruses (ERVs): retroviral sequences that are transmitted vertically as part of the host germ line, within which they may continue to replicate and evolve. ERVs represent both a unique archive of ancient viral sequence information and a dynamic component of host genomes. As such they hold great potential as informative markers for studies of both virus evolution and host genome evolution. Numerous novel ERVs have been described in recent years, particularly as genome sequencing projects have advanced. This review discusses the evolution of ERV lineages, considering the processes by which ERV distribution and diversity is generated. The diversity of ERVs isolated so far is summarised in terms of both their distribution across host taxa, and their relationships to recognised retroviral genera. Finally the relevance of ERVs to studies of genome evolution, host disease and viral ecology is considered, and recent findings discussed.


Assuntos
Retrovirus Endógenos , Evolução Molecular , Variação Genética , Animais , Retrovirus Endógenos/classificação , Retrovirus Endógenos/genética , Retrovirus Endógenos/isolamento & purificação , Humanos , Vertebrados/virologia
20.
Virology ; 311(1): 1-6, 2003 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-12832197

RESUMO

We report the first full-length sequence of an endogenous amphibian retrovirus derived from the African clawed toad Xenopus laevis. The virus, termed Xen1, has one of the largest endogenous retroviral genomes described to date of over 10 kb in length and it also has a relatively complex genomic organisation consisting of LTR-orf1, orf2, gag, pol, env-LTR. Orfs 1 and 2 are novel, duplicated genes of unknown function. Phylogenetic analysis indicates that Xen1 is most closely related to the epsilon -retroviruses WDSV and WEHV types 1 and 2, which are large, complex exogenous retroviruses present within Walleye fish.


Assuntos
Retrovirus Endógenos/genética , Genoma Viral , Provírus/genética , Proteínas Virais/genética , Xenopus laevis/virologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Retrovirus Endógenos/classificação , Dados de Sequência Molecular , Fases de Leitura Aberta , Filogenia , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA