Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Virol ; 87(11): 6137-49, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23514895

RESUMO

Most human immunodeficiency virus (HIV) transmissions are initiated with CCR5 (R5)-using viruses across mucosal surfaces, with the majority in regions where HIV type 1 (HIV-1) clade C predominates. Mucosally transmissible, highly replication competent, pathogenic R5 simian-human immunodeficiency viruses (SHIVs) encoding biologically relevant clade C envelopes are therefore needed as challenge viruses in vaccine efficacy studies with nonhuman primates. Here we describe the generation of three lineage-related subtype C SHIVs through four successive rapid transfers in rhesus macaques of SHIVC109F.PB4, a molecular clone expressing the soluble-CD4 (sCD4)-sensitive CCR5-tropic clade C envelope of a recently infected subject in Zambia. The viruses differed in their monkey passage histories and neutralization sensitivities but remained R5 tropic. SHIVC109P3 and SHIVC109P3N were recovered from a passage-3 rapid-progressor animal during chronic infection (24 weeks postinfection [wpi]) and at end-stage disease (34 wpi), respectively, and are classified as tier 1B strains, whereas SHIVC109P4 was recovered from a passage-4 normal-progressor macaque at 22 wpi and is a tier 2 virus, more difficult to neutralize. All three viruses were transmitted efficiently via intrarectal inoculation, reaching peak viral loads of 10(7) to 10(9) RNA copies/ml plasma and establishing viremia at various set points. Notably, one of seven (GC98) and two of six (CL31, FI08) SHIVC109P3- and SHIVC109P3N-infected macaques, respectively, progressed to AIDS, with neuropathologies observed in GC98 and FI08, as well as coreceptor switching in the latter. These findings support the use of these new SHIVC109F.PB4-derived viruses to study the immunopathology of HIV-1 clade C infection and to evaluate envelope-based AIDS vaccines in nonhuman primates.


Assuntos
Síndrome da Imunodeficiência Adquirida/virologia , HIV-1/genética , HIV-1/fisiologia , Receptores CCR5/metabolismo , Receptores de HIV/metabolismo , Vírus da Imunodeficiência Símia/fisiologia , Síndrome da Imunodeficiência Adquirida/metabolismo , Síndrome da Imunodeficiência Adquirida/patologia , Síndrome da Imunodeficiência Adquirida/transmissão , Sequência de Aminoácidos , Animais , Encéfalo/patologia , Encéfalo/virologia , Modelos Animais de Doenças , Infecções por HIV/imunologia , HIV-1/classificação , Humanos , Macaca mulatta , Dados de Sequência Molecular , Receptores CCR5/genética , Receptores de HIV/genética , Recombinação Genética , Síndrome de Imunodeficiência Adquirida dos Símios , Vírus da Imunodeficiência Símia/classificação , Vírus da Imunodeficiência Símia/genética
2.
Retrovirology ; 10: 9, 2013 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-23369442

RESUMO

BACKGROUND: Mucosally transmissible and pathogenic CCR5 (R5)-tropic simian-human immunodeficiency virus (SHIV) molecular clones are useful reagents to identity neutralization escape in HIV-1 vaccine experiments and to study the envelope evolutionary process and mechanistic basis for coreceptor switch during the course of natural infection. RESULTS: We observed progression to AIDS in rhesus macaques infected intrarectally with molecular clones of the pathogenic R5 SHIVSF162P3N isolate. Expansion to CXCR4 usage was documented in one diseased macaque that mounted a neutralizing antibody response and in another that failed to do so, with the latter displaying a rapid progressor phenotype. V3 loop envelop glycoprotein gp120 sequence changes that are predictive of a CXCR4 (X4)-using phenotype in HIV-1 subtype B primary isolates, specifically basic amino acid substations at positions 11 (S11R), 24 (G24R) and 25 (D25K) of the loop were detected in the two infected macaques. Functional assays showed that envelopes with V3 S11R or D25K mutation were dual-tropic, infecting CD4+ target cells that expressed either the CCR5 or CXCR4 coreceptor. And, consistent with findings of coreceptor switching in macaques infected with the pathogenic isolate, CXCR4-using variant was first detected in the lymph node of the chronically infected rhesus monkey several weeks prior to its presence in peripheral blood. Moreover, X4 emergence in this macaque coincided with persistent peripheral CD4+ T cell loss and a decline in neutralizing antibody titer that are suggestive of immune deterioration, with macrophages as the major virus-producing cells at the end-stage of disease. CONCLUSIONS: The data showed that molecular clones derived from the R5 SHIVSF162P3N isolate are mucosally transmissible and induced disease in a manner similar to that observed in HIV-1 infected individuals, providing a relevant and useful animal infection model for in-depth analyses of host selection pressures and the env evolutionary changes that influence disease outcome, coreceptor switching and vaccine escape.


Assuntos
HIV-1/fisiologia , Mucosa/virologia , Receptores CCR5/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/transmissão , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/fisiologia , Internalização do Vírus , Substituição de Aminoácidos/genética , Animais , Anticorpos Neutralizantes/sangue , Linfócitos T CD4-Positivos/virologia , Anticorpos Anti-HIV/sangue , Proteína gp120 do Envelope de HIV/genética , HIV-1/genética , Macaca mulatta , Receptores de HIV/metabolismo , Recombinação Genética , Reto/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Vírus da Imunodeficiência Símia/genética , Tropismo Viral
3.
J Virol ; 86(17): 9432-42, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22740397

RESUMO

We previously reported efficient transmission of the pathogenic R5 simian-human immunodeficiency virus SHIV(SF162P3N) isolate in Indian rhesus macaques by intravenous and intrarectal inoculations, with a switch to CXCR4 coreceptor usage in ∼50% of infected animals that progressed rapidly to disease. Since women continue to be disproportionately affected by HIV, we developed an animal model based on the intravaginal challenge of female rhesus monkeys with SHIV(SF162P3N) and sought to validate the utility of this model to study relevant aspects of HIV transmission and pathogenesis. The effect of viral dose on infection outcome was evaluated to determine the optimal conditions for the evaluation of HIV-1 preventive and therapeutic strategies. We found that the virus can successfully cross the vaginal mucosal surface to establish infection and induce disease with coreceptor switch, but with lower efficiencies compared to intravenous and rectal transmissions. In contrast to intrarectal infection, peak and cumulative viral load over a 1 year-infection period were significantly greater in macaques exposed intravaginally to lower rather than higher inoculum doses. Moreover, low and transient viremia was observed only in macaques that were challenged intravaginally twice within the same day with a high dose of virus, which can be seen as doubling the dose. Taken together, these results show that SHIV(SF162P3N) can successfully transmit across the genital mucosa, undergo coreceptor switch, and induce disease. However, the administered dose appears to impact SHIV(SF162P3N) vaginal infection outcome in an unexpected manner.


Assuntos
Infecções por HIV/virologia , Receptores CCR5/metabolismo , Receptores de HIV/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/fisiologia , Vagina/virologia , Tropismo Viral , Animais , Modelos Animais de Doenças , Feminino , Infecções por HIV/metabolismo , HIV-1/genética , HIV-1/patogenicidade , HIV-1/fisiologia , Humanos , Macaca mulatta , Mucosa/metabolismo , Mucosa/virologia , Receptores CCR5/genética , Receptores de HIV/genética , Síndrome de Imunodeficiência Adquirida dos Símios/metabolismo , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/patogenicidade , Vagina/metabolismo , Carga Viral , Virulência
4.
J Virol ; 84(24): 12862-71, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20943985

RESUMO

Fitness disadvantage of the transitional intermediates compared to the initial R5 viruses has been suggested to constitute one of the blockades to coreceptor switching, explaining the late appearance of X4 viruses. Using a simian model for human immunodeficiency virus type 1 (HIV-1) coreceptor switching, we demonstrate in this study that similar molecular evolutionary pathways to coreceptor switch occur in more than one R5 simian/human immunodeficiency virus (SHIV)(SF162P3N)-infected macaque. In infected animals where multiple pathways for expansion or switch to CXCR4 coexist, fitness of the transitional intermediates in coreceptor usage efficiency influences their outgrowth and representation in the infecting virus population. Dualtropic and X4 viruses appear at different disease stages, but they have lower entry efficiency than the coexisting R5 strains, which may explain why they do not outcompete the R5 viruses. Similar observations were made in two infected macaques with coreceptor switch, providing in vivo evidence that fitness disadvantage is an obstacle to X4 emergence and expansion.


Assuntos
Infecções por HIV/virologia , HIV/patogenicidade , Glicoproteínas de Membrana/genética , Receptores CCR5/genética , Receptores CXCR4/genética , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/patogenicidade , Proteínas do Envelope Viral/genética , Sequência de Aminoácidos , Animais , DNA Viral/genética , Evolução Molecular , Infecções por HIV/genética , Infecções por HIV/imunologia , Humanos , Macaca mulatta , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Homologia de Sequência de Aminoácidos , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Internalização do Vírus , Replicação Viral
5.
J Virol ; 84(1): 340-51, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19846515

RESUMO

We previously reported coreceptor switch in rhesus macaques inoculated intravenously with R5 simian-human immunodeficiency virus SF162P3N (SHIV(SF162P3N)). Whether R5-to-X4 virus evolution occurs in mucosally infected animals and in which anatomic site the switch occurs, however, were not addressed. We herein report a change in coreceptor preference in macaques infected intrarectally with SHIV(SF162P3N). The switch occurred in infected animals with high levels of virus replication and undetectable antiviral antibody response and required sequence changes in the V3 loop of the gp120 envelope protein. X4 virus emergence was associated with an accelerated drop in peripheral CD4(+) T-cell count but followed rather than preceded the onset of CD4(+) T-cell loss. The conditions, genotypic requirements, and patterns of coreceptor switch in intrarectally infected animals were thus remarkably consistent with those found in macaques infected intravenously. They also overlapped with those reported for humans, suggestive of a common mechanism for coreceptor switch in the two hosts. Furthermore, two independent R5-to-X4 evolutionary pathways were identified in one infected animal, giving rise to dual-tropic and X4 viruses which differed in switch kinetics and tissue localization. The dual-tropic switch event predominated early, and the virus established infection in multiple tissues sites. In contrast, the switch to X4 virus occurred later, initiating and expanding mainly in peripheral lymph nodes. These findings help define R5 SHIV(SF162P3N) infection of rhesus macaques as a model to study the mechanistic basis, dynamics, and sites of HIV-1 coreceptor switch.


Assuntos
Infecções por HIV/virologia , HIV/patogenicidade , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/patogenicidade , Sequência de Aminoácidos , Animais , Antígenos CD4 , HIV/química , Proteína gp120 do Envelope de HIV/química , Humanos , Macaca , Receptores Virais , Vírus da Imunodeficiência Símia/química , Distribuição Tecidual , Replicação Viral
6.
J Transl Med ; 9 Suppl 1: S7, 2011 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-21284906

RESUMO

SIV or SHIV infection of nonhuman primates (NHP) has been used to investigate the impact of coreceptor usage on the composition and dynamics of the CD4+ T cell compartment, mechanisms of disease induction and development of clinical syndrome. As the entire course of infection can be followed, with frequent access to tissue compartments, infection of rhesus macaques with CCR5-tropic SHIVs further allows for study of HIV-1 coreceptor switch after intravenous and mucosal inoculation, with longitudinal and systemic analysis to determine the timing, anatomical sites and cause for the change in envelope glycoprotein and coreceptor preference. Here, we review our current understanding of coreceptor use in NHPs and their impact on the pathobiological characteristics of the infection, and discuss recent advances in NHP studies to uncover the underlying selective pressures for the change in coreceptor preference in vivo.


Assuntos
Linfócitos T CD4-Positivos/imunologia , HIV-1/metabolismo , Receptores CCR5/imunologia , Produtos do Gene env do Vírus da Imunodeficiência Humana/imunologia , Animais , Cercocebus atys , Chlorocebus aethiops , Modelos Animais de Doenças , Humanos , Sistema Imunitário , Mandrillus/metabolismo , Camundongos , Mucosa/virologia , Fenótipo , Receptores CXCR4/química
7.
J Virol ; 82(4): 1899-907, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18077725

RESUMO

Severe acute respiratory syndrome (SARS) is caused by the SARS-associated coronavirus (SARS-CoV), which uses angiotensin-converting enzyme 2 (ACE2) as its receptor for cell entry. A group of SARS-like CoVs (SL-CoVs) has been identified in horseshoe bats. SL-CoVs and SARS-CoVs share identical genome organizations and high sequence identities, with the main exception of the N terminus of the spike protein (S), known to be responsible for receptor binding in CoVs. In this study, we investigated the receptor usage of the SL-CoV S by combining a human immunodeficiency virus-based pseudovirus system with cell lines expressing the ACE2 molecules of human, civet, or horseshoe bat. In addition to full-length S of SL-CoV and SARS-CoV, a series of S chimeras was constructed by inserting different sequences of the SARS-CoV S into the SL-CoV S backbone. Several important observations were made from this study. First, the SL-CoV S was unable to use any of the three ACE2 molecules as its receptor. Second, the SARS-CoV S failed to enter cells expressing the bat ACE2. Third, the chimeric S covering the previously defined receptor-binding domain gained its ability to enter cells via human ACE2, albeit with different efficiencies for different constructs. Fourth, a minimal insert region (amino acids 310 to 518) was found to be sufficient to convert the SL-CoV S from non-ACE2 binding to human ACE2 binding, indicating that the SL-CoV S is largely compatible with SARS-CoV S protein both in structure and in function. The significance of these findings in relation to virus origin, virus recombination, and host switching is discussed.


Assuntos
Quirópteros/virologia , Peptidil Dipeptidase A/metabolismo , Receptores Virais/metabolismo , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , Proteínas Virais/metabolismo , Internalização do Vírus , Sequência de Aminoácidos , Enzima de Conversão de Angiotensina 2 , Animais , Linhagem Celular , Quimera/genética , Quimera/fisiologia , Clonagem Molecular , HIV/genética , Células HeLa , Humanos , Camundongos , Dados de Sequência Molecular , Peptidil Dipeptidase A/análise , Peptidil Dipeptidase A/genética , Mapeamento de Interação de Proteínas , Ratos , Receptores Virais/análise , Receptores Virais/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Montagem de Vírus , Viverridae/virologia
8.
Cell Rep ; 27(9): 2593-2607.e5, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31141685

RESUMO

The HIV-1 envelope glycoprotein (Env) maintains a delicate balance between mediating viral entry and escaping antibody neutralization. Adaptation during transmission of neutralization-sensitive Envs with an "open" conformation remains poorly understood. By passaging a replication-competent simian-human immunodeficiency virus carrying a highly neutralization-sensitive Env (SHIVCNE40) in rhesus macaques, we show that SHIVCNE40 develops enhanced replication kinetics associated with neutralization resistance against antibodies and autologous serum. A gp41 substitution, E658K, functions as the major determinant for these properties. Structural modeling and functional verification indicate that the substitution disrupts an intermolecular salt bridge with the neighboring protomer, thereby promoting fusion and facilitating immune evasion. This effect is applicable across diverse HIV-1 subtypes. Our results highlight the critical role of gp41 in shaping the neutralization profile and the overall conformation of Env during viral adaptation. The unique intermolecular salt bridge could potentially be utilized for rational vaccine design involving more stable HIV-1 envelope trimers.


Assuntos
Adaptação Fisiológica , Anticorpos Neutralizantes/imunologia , Anticorpos Anti-HIV/imunologia , Proteína gp41 do Envelope de HIV/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/imunologia , Internalização do Vírus , Substituição de Aminoácidos , Animais , Feminino , Proteína gp41 do Envelope de HIV/genética , Proteína gp41 do Envelope de HIV/metabolismo , Humanos , Evasão da Resposta Imune , Macaca mulatta , Mutação , Testes de Neutralização , Conformação Proteica , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Vírus da Imunodeficiência Símia/fisiologia
9.
AIDS ; 29(10): F1-8, 2015 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-26035329

RESUMO

OBJECTIVE: Gender differences in immune response and the rate of disease progression in HIV-infected individuals have been reported but the underlying mechanism remains unclear, in part because of the lack of relevant animal models. Here, we report a novel nonhuman primate model for investigation of sex disparity in HIV disease progression. DESIGN/METHODS: Viral load and rate of disease progression were evaluated in rhesus macaques infected intrarectally with lineage-related subtype C R5 simian HIVs. Cytokine/chemokine levels in rectal swab eluates, and bacterial species adherent to the swabs and in the feces were determined. RESULTS: Simian HIV-infected female rhesus macaques progressed faster to AIDS than male macaques, recapitulating the sex bias in HIV-1 disease in humans. There were no significant differences in the levels of soluble immune mediators in the rectal mucosa of naive female and male macaques. However, an exploratory longitudinal study in six infected macaques indicates that the female macaques mounted an earlier and more robust proinflammatory skewed rectal immune response to infection. Moreover, expansion of Proteobacteria that increase in other intestinal inflammatory disorders was significantly higher in the rectal mucosa of female than male macaques during acute infection. CONCLUSION: These findings suggest that sex differences in local innate immune activation and compositional shifts in the gut microbiota could be the drivers of increased disease susceptibility in female macaques. Further studies with this novel nonhuman primate model of HIV infection could lead to innovative research on gender differences, which may have important therapeutic implications for controlling disease in infected men as well as women.


Assuntos
Imunidade Inata , Inflamação/imunologia , Inflamação/patologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Vírus da Imunodeficiência Símia/imunologia , Animais , Biota , Citocinas/análise , Modelos Animais de Doenças , Progressão da Doença , Suscetibilidade a Doenças , Fezes/microbiologia , Feminino , Humanos , Macaca mulatta , Masculino , Reto/microbiologia , Reto/patologia , Fatores Sexuais , Carga Viral
10.
AIDS ; 25(15): 1833-41, 2011 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-21750420

RESUMO

OBJECTIVE: Development of an effective vaccine or topical compound to prevent HIV transmission remains a major goal for control of the AIDS pandemic. Using a nonhuman primate model of heterosexual HIV-1 transmission, we tested whether a topical microbicide that reduces viral infectivity can potentiate the efficacy of a T-cell-based HIV vaccine. DESIGN: A DNA prime and rAd5 virus boost vaccination strategy was employed, and a topical microbicide against the HIV nucleocapsid protein was used. To rigorously test the combination hypothesis, the vaccine constructs contained only two transgenes and the topical microbicide inhibitor was used at a suboptimal dose. Vaccinees were exposed in the absence and presence of the topical microbicide to repeated vaginal R5 simian human immunodeficiency virus (SHIV)(SF162P3) challenge at an escalating dose to more closely mimic high-risk exposure of women to HIV. METHODS: Infection status was determined by PCR. Antiviral immune responses were evaluated by gp120 ELISA and intracellular cytokine staining. RESULTS: A significant delay in SHIV acquisition (log-rank test; P = 0.0416) was seen only in vaccinated macaques that were repeatedly challenged in the presence of the topical microbicide. Peak acute viremia was lower (Mann-Whitney test; P = 0.0387) and viral burden was also reduced (Mann-Whitney test; P = 0.0252) in the combination-treated animals. CONCLUSION: The combined use of a topical microbicide to lower the initial viral seeding/spread and a T-cell-based vaccine to immunologically contain the early virological events of mucosal transmission holds promise as a preventive approach to control the spread of the AIDS epidemic.


Assuntos
Anti-Infecciosos Locais/farmacologia , Anticorpos Antivirais/imunologia , HIV-1 , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Replicação Viral/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Feminino , Macaca mulatta , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Vacinas Virais/farmacologia , Replicação Viral/efeitos dos fármacos
11.
J Gen Virol ; 87(Pt 11): 3355-3359, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17030870

RESUMO

Bats were recently identified as natural reservoirs of SARS-like coronavirus (SL-CoV) or SARS coronavirus-like virus. These viruses, together with SARS coronaviruses (SARS-CoV) isolated from human and palm civet, form a distinctive cluster within the group 2 coronaviruses of the genus Coronavirus, tentatively named group 2b (G2b). In this study, complete genome sequences of two additional group 2b coronaviruses (G2b-CoVs) were determined from horseshoe bat Rhinolophus ferrumequinum (G2b-CoV Rf1) and Rhinolophus macrotis (G2b-CoV Rm1). The bat G2b-CoV isolates have an identical genome organization and share an overall genome sequence identity of 88-92 % among themselves and between them and the human/civet isolates. The most variable regions are located in the genes encoding nsp3, ORF3a, spike protein and ORF8 when bat and human/civet G2b-CoV isolates are compared. Genetic analysis demonstrated that a diverse G2b-CoV population exists in the bat habitat and has evolved from a common ancestor of SARS-CoV.


Assuntos
Quirópteros/virologia , Genoma Viral , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Animais , China , Variação Genética , Hemaglutininas Virais/genética , Humanos , Glicoproteínas de Membrana/genética , Fases de Leitura Aberta/genética , RNA Polimerase Dependente de RNA/genética , Homologia de Sequência de Aminoácidos , Síndrome Respiratória Aguda Grave/virologia , Glicoproteína da Espícula de Coronavírus , Proteínas do Envelope Viral/genética , Proteínas da Matriz Viral/genética , Proteínas não Estruturais Virais/genética , Proteínas Virais/genética , Viverridae/virologia
12.
Science ; 310(5748): 676-9, 2005 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-16195424

RESUMO

Severe acute respiratory syndrome (SARS) emerged in 2002 to 2003 in southern China. The origin of its etiological agent, the SARS coronavirus (SARS-CoV), remains elusive. Here we report that species of bats are a natural host of coronaviruses closely related to those responsible for the SARS outbreak. These viruses, termed SARS-like coronaviruses (SL-CoVs), display greater genetic variation than SARS-CoV isolated from humans or from civets. The human and civet isolates of SARS-CoV nestle phylogenetically within the spectrum of SL-CoVs, indicating that the virus responsible for the SARS outbreak was a member of this coronavirus group.


Assuntos
Quirópteros/virologia , Coronavirus , Reservatórios de Doenças , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave , Sequência de Aminoácidos , Animais , China/epidemiologia , Chlorocebus aethiops , Doenças Transmissíveis Emergentes , Coronavirus/classificação , Surtos de Doenças , Variação Genética , Genoma Viral , Henipavirus/classificação , Humanos , Dados de Sequência Molecular , Mutação , Filogenia , Reação em Cadeia da Polimerase , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/classificação , Análise de Sequência de DNA , Síndrome Respiratória Aguda Grave/epidemiologia , Síndrome Respiratória Aguda Grave/transmissão , Síndrome Respiratória Aguda Grave/virologia , Células Vero , Viverridae/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA