Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Emerg Infect Dis ; 24(7): 1128-1238, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29912683

RESUMO

Influenza viruses exist in each host as a collection of genetically diverse variants, which might enhance their adaptive potential. To assess the genetic and functional diversity of highly pathogenic avian influenza A(H5N1) viruses within infected humans, we used deep-sequencing methods to characterize samples obtained from infected patients in northern Vietnam during 2004-2010 on different days after infection, from different anatomic sites, or both. We detected changes in virus genes that affected receptor binding, polymerase activity, or interferon antagonism, suggesting that these factors could play roles in influenza virus adaptation to humans. However, the frequency of most of these mutations remained low in the samples tested, implying that they were not efficiently selected within these hosts. Our data suggest that adaptation of influenza A(H5N1) viruses is probably stepwise and depends on accumulating combinations of mutations that alter function while maintaining fitness.


Assuntos
Variação Genética , Virus da Influenza A Subtipo H5N1/classificação , Virus da Influenza A Subtipo H5N1/genética , Influenza Humana/epidemiologia , Influenza Humana/virologia , Animais , Linhagem Celular , Genes Virais , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , História do Século XXI , Humanos , Influenza Humana/história , Tipagem Molecular , Filogenia , Vigilância da População , Vietnã/epidemiologia , Tropismo Viral
2.
J Virol ; 91(4)2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-27974564

RESUMO

Simian arteriviruses are a diverse clade of viruses infecting captive and wild nonhuman primates. We recently reported that Kibale red colobus virus 1 (KRCV-1) causes a mild and self-limiting disease in experimentally infected crab-eating macaques, while simian hemorrhagic fever virus (SHFV) causes lethal viral hemorrhagic fever. Here we characterize how these viruses evolved during replication in cell culture and in experimentally infected macaques. During passage in cell culture, 68 substitutions that were localized in open reading frames (ORFs) likely associated with host cell entry and exit became fixed in the KRCV-1 genome. However, we did not detect any strong signatures of selection during replication in macaques. We uncovered patterns of evolution that were distinct from those observed in surveys of wild red colobus monkeys, suggesting that these species may exert different adaptive challenges for KRCV-1. During SHFV infection, we detected signatures of selection on ORF 5a and on a small subset of sites in the genome. Overall, our data suggest that patterns of evolution differ markedly among simian arteriviruses and among host species. IMPORTANCE: Certain RNA viruses can cross species barriers and cause disease in new hosts. Simian arteriviruses are a diverse group of related viruses that infect captive and wild nonhuman primates, with associated disease severity ranging from apparently asymptomatic infections to severe, viral hemorrhagic fevers. We infected nonhuman primate cell cultures and then crab-eating macaques with either simian hemorrhagic fever virus (SHFV) or Kibale red colobus virus 1 (KRCV-1) and assessed within-host viral evolution. We found that KRCV-1 quickly acquired a large number of substitutions in its genome during replication in cell culture but that evolution in macaques was limited. In contrast, we detected selection focused on SHFV ORFs 5a and 5, which encode putative membrane proteins. These patterns suggest that in addition to diverse pathogenic phenotypes, these viruses may also exhibit distinct patterns of within-host evolution both in vitro and in vivo.


Assuntos
Infecções por Arterivirus/veterinária , Arterivirus/fisiologia , Evolução Biológica , Interações Hospedeiro-Patógeno , Doenças dos Macacos/virologia , Animais , Interações Hospedeiro-Patógeno/genética , Macaca fascicularis , Doenças dos Macacos/genética , Fases de Leitura Aberta , Polimorfismo de Nucleotídeo Único , RNA Viral , Seleção Genética , Internalização do Vírus , Replicação Viral
3.
PLoS Pathog ; 12(12): e1006048, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27926931

RESUMO

Within the first three weeks of human immunodeficiency virus (HIV) infection, virus replication peaks in peripheral blood. Despite the critical, causal role of virus replication in determining transmissibility and kinetics of progression to acquired immune deficiency syndrome (AIDS), there is limited understanding of the conditions required to transform the small localized transmitted founder virus population into a large and heterogeneous systemic infection. Here we show that during the hyperacute "pre-peak" phase of simian immunodeficiency virus (SIV) infection in macaques, high levels of microbial DNA transiently translocate into peripheral blood. This, heretofore unappreciated, hyperacute-phase microbial translocation was accompanied by sustained reduction of lipopolysaccharide (LPS)-specific antibody titer, intestinal permeability, increased abundance of CD4+CCR5+ T cell targets of virus replication, and T cell activation. To test whether increasing gastrointestinal permeability to cause microbial translocation would amplify viremia, we treated two SIV-infected macaque 'elite controllers' with a short-course of dextran sulfate sodium (DSS)-stimulating a transient increase in microbial translocation and a prolonged recrudescent viremia. Altogether, our data implicates translocating microbes as amplifiers of immunodeficiency virus replication that effectively undermine the host's capacity to contain infection.


Assuntos
DNA Viral/sangue , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/patogenicidade , Viremia/virologia , Animais , Progressão da Doença , Feminino , Citometria de Fluxo , Imunofenotipagem , Inflamação/imunologia , Inflamação/virologia , Ativação Linfocitária/imunologia , Macaca fascicularis , Masculino , Reação em Cadeia da Polimerase , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Replicação Viral/imunologia
4.
Proc Natl Acad Sci U S A ; 112(44): 13645-50, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26483473

RESUMO

Elucidation of maternal immune correlates of protection against congenital cytomegalovirus (CMV) is necessary to inform future vaccine design. Here, we present a novel rhesus macaque model of placental rhesus CMV (rhCMV) transmission and use it to dissect determinants of protection against congenital transmission following primary maternal rhCMV infection. In this model, asymptomatic intrauterine infection was observed following i.v. rhCMV inoculation during the early second trimester in two of three rhCMV-seronegative pregnant females. In contrast, fetal loss or infant CMV-associated sequelae occurred in four rhCMV-seronegative pregnant macaques that were CD4(+) T-cell depleted at the time of inoculation. Animals that received the CD4(+) T-cell-depleting antibody also exhibited higher plasma and amniotic fluid viral loads, dampened virus-specific CD8(+) T-cell responses, and delayed production of autologous neutralizing antibodies compared with immunocompetent monkeys. Thus, maternal CD4(+) T-cell immunity during primary rhCMV infection is important for controlling maternal viremia and inducing protective immune responses that prevent severe CMV-associated fetal disease.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Infecções por Citomegalovirus/prevenção & controle , Transmissão Vertical de Doenças Infecciosas , Troca Materno-Fetal , Animais , Anticorpos Antivirais/imunologia , Infecções por Citomegalovirus/congênito , Infecções por Citomegalovirus/transmissão , Modelos Animais de Doenças , Feminino , Macaca mulatta , Gravidez
5.
J Virol ; 90(2): 630-5, 2016 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-26559828

RESUMO

Wild nonhuman primates are immediate sources and long-term reservoirs of human pathogens. However, ethical and technical challenges have hampered the identification of novel blood-borne pathogens in these animals. We recently examined RNA viruses in plasma from wild African monkeys and discovered several novel, highly divergent viruses belonging to the family Arteriviridae. Close relatives of these viruses, including simian hemorrhagic fever virus, have caused sporadic outbreaks of viral hemorrhagic fever in captive macaque monkeys since the 1960s. However, arterivirus infection in wild nonhuman primates had not been described prior to 2011. The arteriviruses recently identified in wild monkeys have high sequence and host species diversity, maintain high viremia, and are prevalent in affected populations. Taken together, these features suggest that the simian arteriviruses may be "preemergent" zoonotic pathogens. If not, this would imply that biological characteristics of RNA viruses thought to facilitate zoonotic transmission may not, by themselves, be sufficient for such transmission to occur.


Assuntos
Infecções por Arterivirus/transmissão , Infecções por Arterivirus/veterinária , Arterivirus/fisiologia , Doenças dos Primatas/transmissão , Doenças dos Primatas/virologia , Zoonoses/transmissão , Zoonoses/virologia , Animais , Arterivirus/genética , Infecções por Arterivirus/virologia , Haplorrinos , Humanos
6.
J Virol ; 90(15): 6724-6737, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27170760

RESUMO

UNLABELLED: Nonhuman primates (NHPs) are a historically important source of zoonotic viruses and are a gold-standard model for research on many human pathogens. However, with the exception of simian immunodeficiency virus (SIV) (family Retroviridae), the blood-borne viruses harbored by these animals in the wild remain incompletely characterized. Here, we report the discovery and characterization of two novel simian pegiviruses (family Flaviviridae) and two novel simian arteriviruses (family Arteriviridae) in wild African green monkeys from Zambia (malbroucks [Chlorocebus cynosuros]) and South Africa (vervet monkeys [Chlorocebus pygerythrus]). We examine several aspects of infection, including viral load, genetic diversity, evolution, and geographic distribution, as well as host factors such as age, sex, and plasma cytokines. In combination with previous efforts to characterize blood-borne RNA viruses in wild primates across sub-Saharan Africa, these discoveries demonstrate that in addition to SIV, simian pegiviruses and simian arteriviruses are widespread and prevalent among many African cercopithecoid (i.e., Old World) monkeys. IMPORTANCE: Primates are an important source of viruses that infect humans and serve as an important laboratory model of human virus infection. Here, we discover two new viruses in African green monkeys from Zambia and South Africa. In combination with previous virus discovery efforts, this finding suggests that these virus types are widespread among African monkeys. Our analysis suggests that one of these virus types, the simian arteriviruses, may have the potential to jump between different primate species and cause disease. In contrast, the other virus type, the pegiviruses, are thought to reduce the disease caused by human immunodeficiency virus (HIV) in humans. However, we did not observe a similar protective effect in SIV-infected African monkeys coinfected with pegiviruses, possibly because SIV causes little to no disease in these hosts.


Assuntos
Infecções por Arterivirus/epidemiologia , Evolução Biológica , Infecções por Flaviviridae/epidemiologia , Variação Genética , Infecções por Lentivirus/epidemiologia , Carga Viral , África/epidemiologia , Animais , Animais Selvagens , Arterivirus/genética , Arterivirus/patogenicidade , Infecções por Arterivirus/genética , Infecções por Arterivirus/virologia , Flaviviridae/genética , Flaviviridae/patogenicidade , Infecções por Flaviviridae/genética , Infecções por Flaviviridae/virologia , Genoma Viral , Haplorrinos , Humanos , Lentivirus/genética , Lentivirus/patogenicidade , Infecções por Lentivirus/genética , Infecções por Lentivirus/virologia , Filogenia , Prevalência
8.
J Virol ; 89(4): 2425-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25473056

RESUMO

In 49 patients with known Ebola virus disease outcomes during the ongoing outbreak in Sierra Leone, 13 were coinfected with the immunomodulatory pegivirus GB virus C (GBV-C). Fifty-three percent of these GBV-C(+) patients survived; in contrast, only 22% of GBV-C(-) patients survived. Both survival and GBV-C status were associated with age, with older patients having lower survival rates and intermediate-age patients (21 to 45 years) having the highest rate of GBV-C infection. Understanding the separate and combined effects of GBV-C and age on Ebola virus survival may lead to new treatment and prevention strategies, perhaps through age-related pathways of immune activation.


Assuntos
Coinfecção/epidemiologia , Infecções por Flaviviridae/epidemiologia , Vírus GB C/isolamento & purificação , Doença pelo Vírus Ebola/complicações , Hepatite Viral Humana/epidemiologia , Adolescente , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Criança , Pré-Escolar , Coinfecção/virologia , Feminino , Infecções por Flaviviridae/virologia , Vírus GB C/genética , Doença pelo Vírus Ebola/mortalidade , Hepatite Viral Humana/virologia , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , RNA Viral/genética , Análise de Sequência de DNA , Serra Leoa/epidemiologia , Análise de Sobrevida , Adulto Jovem
9.
J Virol ; 89(15): 8082-7, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25972539

RESUMO

Simian hemorrhagic fever (SHF) is lethal for macaques. Based on clinical presentation and serological diagnosis, all reported SHF outbreaks were thought to be caused by different strains of the same virus, simian hemorrhagic fever virus (SHFV; Arteriviridae). Here we show that the SHF outbreaks in Sukhumi in 1964 and in Alamogordo in 1989 were caused not by SHFV but by two novel divergent arteriviruses. Our results indicate that multiple divergent simian arteriviruses can cause SHF.


Assuntos
Infecções por Arterivirus/veterinária , Arterivirus/isolamento & purificação , Febres Hemorrágicas Virais/veterinária , Macaca/virologia , Doenças dos Primatas/virologia , Sequência de Aminoácidos , Animais , Arterivirus/classificação , Arterivirus/genética , Arterivirus/fisiologia , Infecções por Arterivirus/história , Infecções por Arterivirus/virologia , Evolução Molecular , Febres Hemorrágicas Virais/história , Febres Hemorrágicas Virais/virologia , História do Século XX , Humanos , Dados de Sequência Molecular , Filogenia , Doenças dos Primatas/história , Homologia de Sequência de Aminoácidos , Proteínas Virais/química , Proteínas Virais/genética
10.
J Virol ; 89(1): 844-56, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25355889

RESUMO

UNLABELLED: Simian hemorrhagic fever virus (SHFV) causes a severe and almost uniformly fatal viral hemorrhagic fever in Asian macaques but is thought to be nonpathogenic for humans. To date, the SHFV life cycle is almost completely uncharacterized on the molecular level. Here, we describe the first steps of the SHFV life cycle. Our experiments indicate that SHFV enters target cells by low-pH-dependent endocytosis. Dynamin inhibitors, chlorpromazine, methyl-ß-cyclodextrin, chloroquine, and concanamycin A dramatically reduced SHFV entry efficiency, whereas the macropinocytosis inhibitors EIPA, blebbistatin, and wortmannin and the caveolin-mediated endocytosis inhibitors nystatin and filipin III had no effect. Furthermore, overexpression and knockout study and electron microscopy results indicate that SHFV entry occurs by a dynamin-dependent clathrin-mediated endocytosis-like pathway. Experiments utilizing latrunculin B, cytochalasin B, and cytochalasin D indicate that SHFV does not hijack the actin polymerization pathway. Treatment of target cells with proteases (proteinase K, papain, α-chymotrypsin, and trypsin) abrogated entry, indicating that the SHFV cell surface receptor is a protein. Phospholipases A2 and D had no effect on SHFV entry. Finally, treatment of cells with antibodies targeting CD163, a cell surface molecule identified as an entry factor for the SHFV-related porcine reproductive and respiratory syndrome virus, diminished SHFV replication, identifying CD163 as an important SHFV entry component. IMPORTANCE: Simian hemorrhagic fever virus (SHFV) causes highly lethal disease in Asian macaques resembling human illness caused by Ebola or Lassa virus. However, little is known about SHFV's ecology and molecular biology and the mechanism by which it causes disease. The results of this study shed light on how SHFV enters its target cells. Using electron microscopy and inhibitors for various cellular pathways, we demonstrate that SHFV invades cells by low-pH-dependent, actin-independent endocytosis, likely with the help of a cellular surface protein.


Assuntos
Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Arterivirus/fisiologia , Endocitose , Interações Hospedeiro-Patógeno , Receptores de Superfície Celular/metabolismo , Receptores Virais/metabolismo , Internalização do Vírus , Animais , Linhagem Celular , Chlorocebus aethiops
11.
Arch Virol ; 161(3): 755-68, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26608064

RESUMO

The family Arteriviridae presently includes a single genus Arterivirus. This genus includes four species as the taxonomic homes for equine arteritis virus (EAV), lactate dehydrogenase-elevating virus (LDV), porcine respiratory and reproductive syndrome virus (PRRSV), and simian hemorrhagic fever virus (SHFV), respectively. A revision of this classification is urgently needed to accommodate the recent description of eleven highly divergent simian arteriviruses in diverse African nonhuman primates, one novel arterivirus in an African forest giant pouched rat, and a novel arterivirus in common brushtails in New Zealand. In addition, the current arterivirus nomenclature is not in accordance with the most recent version of the International Code of Virus Classification and Nomenclature. Here we outline an updated, amended, and improved arterivirus taxonomy based on current data. Taxon-specific sequence cut-offs are established relying on a newly established open reading frame 1b phylogeny and pairwise sequence comparison (PASC) of coding-complete arterivirus genomes. As a result, the current genus Arterivirus is replaced by five genera: Equartevirus (for EAV), Rodartevirus (LDV + PRRSV), Simartevirus (SHFV + simian arteriviruses), Nesartevirus (for the arterivirus from forest giant pouched rats), and Dipartevirus (common brushtail arterivirus). The current species Porcine reproductive and respiratory syndrome virus is divided into two species to accommodate the clear divergence of the European and American "types" of PRRSV, both of which now receive virus status. The current species Simian hemorrhagic fever virus is divided into nine species to accommodate the twelve known simian arteriviruses. Non-Latinized binomial species names are introduced to replace all current species names to clearly differentiate them from virus names, which remain largely unchanged.


Assuntos
Arteriviridae/classificação , Arteriviridae/isolamento & purificação , Infecções por Vírus de RNA/veterinária , Arteriviridae/genética , Análise por Conglomerados , Genoma Viral , Fases de Leitura Aberta , Filogenia , RNA Viral/genética , Homologia de Sequência , Terminologia como Assunto
12.
J Virol ; 88(22): 13231-9, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25187550

RESUMO

UNLABELLED: Since the 1960s, simian hemorrhagic fever virus (SHFV; Nidovirales, Arteriviridae) has caused highly fatal outbreaks of viral hemorrhagic fever in captive Asian macaque colonies. However, the source(s) of these outbreaks and the natural reservoir(s) of this virus remain obscure. Here we report the identification of two novel, highly divergent simian arteriviruses related to SHFV, Mikumi yellow baboon virus 1 (MYBV-1) and Southwest baboon virus 1 (SWBV-1), in wild and captive baboons, respectively, and demonstrate the recent transmission of SWBV-1 among captive baboons. These findings extend our knowledge of the genetic and geographic diversity of the simian arteriviruses, identify baboons as a natural host of these viruses, and provide further evidence that baboons may have played a role in previous outbreaks of simian hemorrhagic fever in macaques, as has long been suspected. This knowledge should aid in the prevention of disease outbreaks in captive macaques and supports the growing body of evidence that suggests that simian arterivirus infections are common in Old World monkeys of many different species throughout Africa. IMPORTANCE: Historically, the emergence of primate viruses both in humans and in other primate species has caused devastating outbreaks of disease. One strategy for preventing the emergence of novel primate pathogens is to identify microbes with the potential for cross-species transmission in their natural state within reservoir species from which they might emerge. Here, we detail the discovery and characterization of two related simian members of the Arteriviridae family that have a history of disease emergence and host switching. Our results expand the phylogenetic and geographic range of the simian arteriviruses and define baboons as a natural host for these viruses. Our findings also identify a potential threat to captive macaque colonies by showing that simian arteriviruses are actively circulating in captive baboons.


Assuntos
Arteriviridae/classificação , Arteriviridae/isolamento & purificação , Doenças dos Macacos/virologia , Infecções por Vírus de RNA/veterinária , Animais , Animais Selvagens , Animais de Zoológico , Arteriviridae/genética , Feminino , Variação Genética , Masculino , Dados de Sequência Molecular , Papio , Filogeografia , Infecções por Vírus de RNA/virologia , RNA Viral/genética , Análise de Sequência de DNA , Topografia Médica
13.
Emerg Infect Dis ; 20(1): 109-13, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24377497

RESUMO

A captive juvenile Bornean orangutan (Pongo pygmaeus) died from an unknown disseminated parasitic infection. Deep sequencing of DNA from infected tissues, followed by gene-specific PCR and sequencing, revealed a divergent species within the newly proposed genus Versteria (Cestoda: Taeniidae). Versteria may represent a previously unrecognized risk to primate health.


Assuntos
Doenças dos Símios Antropoides/parasitologia , Cestoides/classificação , Cestoides/genética , Infecções por Cestoides/veterinária , Pongo pygmaeus/parasitologia , Animais , Doenças dos Símios Antropoides/patologia , Genes de Helmintos , Filogenia , RNA Ribossômico
14.
Retrovirology ; 11: 55, 2014 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-24996566

RESUMO

BACKGROUND: Human immunodeficiency virus (HIV) type 1 and 2, the causative agents of acquired immunodeficiency syndrome (AIDS), emerged from African non-human primates (NHPs) through zoonotic transmission of simian immunodeficiency viruses (SIV). Among African NHPs, the Cercopithecus genus contains the largest number of species known to harbor SIV. However, our understanding of the diversity and evolution of SIVs infecting this genus is limited by incomplete taxonomic and geographic sampling, particularly in East Africa. In this study, we screened blood specimens from red-tailed guenons (Cercopithecus ascanius schmidti) from Kibale National Park, Uganda, for the presence of novel SIVs using unbiased deep-sequencing. FINDINGS: We describe and characterize the first full-length SIV genomes from wild red-tailed guenons in Kibale National Park, Uganda. This new virus, tentatively named SIVrtg_Kib, was detected in five out of twelve animals and is highly divergent from other Cercopithecus SIVs as well as from previously identified SIVs infecting red-tailed guenons, thus forming a new SIV lineage. CONCLUSIONS: Our results show that the genetic diversity of SIVs infecting red-tailed guenons is greater than previously appreciated. This diversity could be the result of cross-species transmission between different guenon species or limited gene flow due to geographic separation among guenon populations.


Assuntos
Cercopithecus/virologia , Genoma Viral , Vírus da Imunodeficiência Símia/genética , Animais , Vírus da Imunodeficiência Símia/classificação , Uganda
15.
J Virol ; 87(1): 688-91, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23077302

RESUMO

Simian hemorrhagic fever virus (SHFV) is an arterivirus that causes severe disease in captive macaques. We describe two new SHFV variants subclinically infecting wild African red-tailed guenons (Cercopithecus ascanius). Both variants are highly divergent from the prototype virus and variants infecting sympatric red colobus (Procolobus rufomitratus). All known SHFV variants are monophyletic and share three open reading frames not present in other arteriviruses. Our data suggest a need to modify the current arterivirus classification.


Assuntos
Infecções por Arterivirus/veterinária , Arterivirus/classificação , Arterivirus/isolamento & purificação , Variação Genética , Doenças dos Primatas/virologia , RNA Viral/genética , África , Animais , Arterivirus/genética , Infecções por Arterivirus/virologia , Infecções Assintomáticas , Cercopithecus , Análise por Conglomerados , Genótipo , Dados de Sequência Molecular , Filogenia , Análise de Sequência de DNA
16.
J Virol ; 87(16): 8971-81, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23740998

RESUMO

GB virus B (GBV-B; family Flaviviridae, genus Hepacivirus) has been studied in New World primates as a model for human hepatitis C virus infection, but the distribution of GBV-B and its relatives in nature has remained obscure. Here, we report the discovery of a novel and highly divergent GBV-B-like virus in an Old World monkey, the black-and-white colobus (Colobus guereza), in Uganda. The new virus, guereza hepacivirus (GHV), clusters phylogenetically with GBV-B and recently described hepaciviruses infecting African bats and North American rodents, and it shows evidence of ancient recombination with these other hepaciviruses. Direct sequencing of reverse-transcribed RNA from blood plasma from three of nine colobus monkeys yielded near-complete GHV genomes, comprising two distinct viral variants. The viruses contain an exceptionally long nonstructural 5A (NS5A) gene, approximately half of which codes for a protein with no discernible homology to known proteins. Computational structure-based analyses indicate that the amino terminus of the GHV NS5A protein may serve a zinc-binding function, similar to the NS5A of other viruses within the family Flaviviridae. However, the 521-amino-acid carboxy terminus is intrinsically disordered, reflecting an unusual degree of structural plasticity and polyfunctionality. These findings shed new light on the natural history and evolution of the hepaciviruses and on the extent of structural variation within the Flaviviridae.


Assuntos
Vírus GB B/genética , Vírus GB B/isolamento & purificação , Hepatite C/veterinária , Doenças dos Primatas/virologia , Proteínas não Estruturais Virais/genética , Animais , Análise por Conglomerados , Colobus , Simulação por Computador , Vírus GB B/química , Genoma Viral , Hepatite C/virologia , Modelos Moleculares , Dados de Sequência Molecular , Filogenia , Conformação Proteica , RNA Viral/genética , Análise de Sequência de DNA , Uganda , Proteínas não Estruturais Virais/química
17.
Retrovirology ; 10: 107, 2013 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-24139306

RESUMO

BACKGROUND: African non-human primates (NHPs) are natural hosts for simian immunodeficiency viruses (SIV), the zoonotic transmission of which led to the emergence of HIV-1 and HIV-2. However, our understanding of SIV diversity and evolution is limited by incomplete taxonomic and geographic sampling of NHPs, particularly in East Africa. In this study, we screened blood specimens from nine black-and-white colobus monkeys (Colobus guereza occidentalis) from Kibale National Park, Uganda, for novel SIVs using a combination of serology and "unbiased" deep-sequencing, a method that does not rely on genetic similarity to previously characterized viruses. RESULTS: We identified two novel and divergent SIVs, tentatively named SIVkcol-1 and SIVkcol-2, and assembled genomes covering the entire coding region for each virus. SIVkcol-1 and SIVkcol-2 were detected in three and four animals, respectively, but with no animals co-infected. Phylogenetic analyses showed that SIVkcol-1 and SIVkcol-2 form a lineage with SIVcol, previously discovered in black-and-white colobus from Cameroon. Although SIVkcol-1 and SIVkcol-2 were isolated from the same host population in Uganda, SIVkcol-1 is more closely related to SIVcol than to SIVkcol-2. Analysis of functional motifs in the extracellular envelope glycoprotein (gp120) revealed that SIVkcol-2 is unique among primate lentiviruses in containing only 16 conserved cysteine residues instead of the usual 18 or more. CONCLUSIONS: Our results demonstrate that the genetic diversity of SIVs infecting black-and-white colobus across equatorial Africa is greater than previously appreciated and that divergent SIVs can co-circulate in the same colobine population. We also show that the use of "unbiased" deep sequencing for the detection of SIV has great advantages over traditional serological approaches, especially for studies of unknown or poorly characterized viruses. Finally, the detection of the first SIV containing only 16 conserved cysteines in the extracellular envelope protein gp120 further expands the range of functional motifs observed among SIVs and highlights the complex evolutionary history of simian retroviruses.


Assuntos
Genoma Viral , RNA Viral/genética , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/isolamento & purificação , Animais , Análise por Conglomerados , Colobus , Variação Genética , Sequenciamento de Nucleotídeos em Larga Escala , Dados de Sequência Molecular , Filogenia , Homologia de Sequência , Vírus da Imunodeficiência Símia/classificação , Uganda
18.
J Gen Virol ; 94(Pt 12): 2670-2678, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24077364

RESUMO

Human pegivirus (HPgV), formerly 'GB virus C' or 'hepatitis G virus', is a member of the genus Flavivirus (Flaviviridae) that has garnered significant attention due to its inhibition of HIV, including slowing disease progression and prolonging survival in HIV-infected patients. Currently, there are six proposed HPgV genotypes that have roughly distinct geographical distributions. Genotypes 2 and 3 are the most comprehensively characterized, whereas those genotypes occurring on the African continent, where HPgV prevalence is highest, are less well studied. Using deep sequencing methods, we identified complete coding HPgV sequences in four of 28 patients (14.3%) in rural Uganda, east Africa. One of these sequences corresponds to genotype 1 and is the first complete genome of this genotype from east Africa. The remaining three sequences correspond to genotype 5, a genotype that was previously considered exclusively South African. All four positive samples were collected within a geographical area of less than 25 km(2), showing that multiple HPgV genotypes co-circulate in this area. Analysis of intra-host viral genetic diversity revealed that total single-nucleotide polymorphism frequency was approximately tenfold lower in HPgV than in hepatitis C virus. Finally, one patient was co-infected with HPgV and HIV, which, in combination with the high prevalence of HIV, suggests that this region would be a useful locale to study the interactions and co-evolution of these viruses.


Assuntos
Infecções por Flaviviridae/epidemiologia , Vírus GB C/genética , Variação Genética , Hepatite Viral Humana/epidemiologia , Sequenciamento de Nucleotídeos em Larga Escala/métodos , População Rural/estatística & dados numéricos , Adolescente , Adulto , Animais , Feminino , Infecções por Flaviviridae/virologia , Vírus GB C/classificação , Genoma Viral , Genótipo , Infecções por HIV/complicações , Infecções por HIV/epidemiologia , Infecções por HIV/virologia , Hepatite Viral Humana/virologia , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Análise de Sequência de DNA , Uganda/epidemiologia , Adulto Jovem
19.
J Virol ; 86(7): 3952-60, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22278255

RESUMO

Hepatitis C virus (HCV) is the leading cause of liver disease worldwide. In this study, we analyzed four treatment-naïve patients infected with subtype 1a and performed Roche/454 pyrosequencing across the coding region. We report the presence of low-level drug resistance mutations that would most likely have been missed using conventional sequencing methods. The approach described here is broadly applicable to studies of viral diversity and could help to improve the efficacy of direct-acting antiviral agents (DAA) in the treatment of HCV-infected patients.


Assuntos
Hepacivirus/genética , Hepatite C/virologia , Especificidade de Hospedeiro , Fases de Leitura Aberta , Análise de Sequência de DNA/métodos , Antivirais/farmacologia , Farmacorresistência Viral , Hepacivirus/classificação , Hepacivirus/efeitos dos fármacos , Hepacivirus/fisiologia , Humanos , Dados de Sequência Molecular , Filogenia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA