Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Clin Infect Dis ; 75(1): e974-e990, 2022 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-35178548

RESUMO

BACKGROUND: The role of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the pathogenesis of testicular damage is uncertain. METHODS: We investigated the virological, pathological, and immunological changes in testes of hamsters challenged by wild-type SARS-CoV-2 and its variants with intranasal or direct testicular inoculation using influenza virus A(H1N1)pdm09 as control. RESULTS: Besides self-limiting respiratory tract infection, intranasal SARS-CoV-2 challenge caused acute decrease in sperm count, serum testosterone and inhibin B at 4-7 days after infection; and chronic reduction in testicular size and weight, and serum sex hormone at 42-120 days after infection. Acute histopathological damage with worsening degree of testicular inflammation, hemorrhage, necrosis, degeneration of seminiferous tubules, and disruption of orderly spermatogenesis were seen with increasing virus inoculum. Degeneration and death of Sertoli and Leydig cells were found. Although viral loads and SARS-CoV-2 nucleocapsid protein expression were markedly lower in testicular than in lung tissues, direct intratesticular injection of SARS-CoV-2 demonstrated nucleocapsid expressing interstitial cells and epididymal epithelial cells, While intranasal or intratesticular challenge by A(H1N1)pdm09 control showed no testicular infection or damage. From 7 to 120 days after infection, degeneration and apoptosis of seminiferous tubules, immune complex deposition, and depletion of spermatogenic cell and spermatozoa persisted. Intranasal challenge with Omicron and Delta variants could also induce similar testicular changes. This testicular damage can be prevented by vaccination. CONCLUSIONS: SARS-CoV-2 can cause acute testicular damage with subsequent chronic asymmetric testicular atrophy and associated hormonal changes despite a self-limiting pneumonia in hamsters. Awareness of possible hypogonadism and subfertility is important in managing convalescent coronavirus disease 2019 in men.


Assuntos
COVID-19 , Vírus da Influenza A Subtipo H1N1 , Animais , Cricetinae , Humanos , Masculino , SARS-CoV-2 , Sêmen , Testículo
2.
Proc Natl Acad Sci U S A ; 115(26): 6822-6827, 2018 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-29891677

RESUMO

Novel reassortant avian influenza H7N9 virus and pandemic 2009 H1N1 (H1N1pdm) virus cause human infections, while avian H7N2 and swine H1N1 virus mainly infect birds and pigs, respectively. There is no robust in vitro model for assessing the infectivity of emerging viruses in humans. Based on a recently established method, we generated long-term expanding 3D human airway organoids which accommodate four types of airway epithelial cells: ciliated, goblet, club, and basal cells. We report differentiation conditions which increase ciliated cell numbers to a nearly physiological level with synchronously beating cilia readily discernible in every organoid. In addition, the differentiation conditions induce elevated levels of serine proteases, which are essential for productive infection of human influenza viruses and low-pathogenic avian influenza viruses. We also established improved 2D monolayer culture conditions for the differentiated airway organoids. To demonstrate the ability of differentiated airway organoids to identify human-infective virus, 3D and 2D differentiated airway organoids are applied to evaluate two pairs of viruses with known distinct infectivity in humans, H7N9/Ah versus H7N2 and H1N1pdm versus an H1N1 strain isolated from swine (H1N1sw). The human-infective H7N9/Ah virus replicated more robustly than the poorly human-infective H7N2 virus; the highly human-infective H1N1pdm virus replicated to a higher titer than the counterpart H1N1sw. Collectively, we developed differentiated human airway organoids which can morphologically and functionally simulate human airway epithelium. These differentiated airway organoids can be applied for rapid assessment of the infectivity of emerging respiratory viruses to human.


Assuntos
Vírus da Influenza A Subtipo H1N1/patogenicidade , Vírus da Influenza A Subtipo H7N2/patogenicidade , Influenza Humana , Organoides/virologia , Sistema Respiratório/virologia , Humanos , Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H7N2/crescimento & desenvolvimento , Organoides/patologia , Sistema Respiratório/patologia
3.
J Infect Dis ; 221(4): 647-659, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-31562757

RESUMO

BACKGROUND: Human infection with Middle East respiratory syndrome coronavirus (MERS-CoV) poses an ongoing threat to public health worldwide. The studies of MERS patients with severe disease and experimentally infected animals showed that robust viral replication and intensive proinflammatory response in lung tissues contribute to high pathogenicity of MERS-CoV. We sought to identify pattern recognition receptor (PRR) signaling pathway(s) that mediates the inflammatory cascade in human macrophages upon MERS-CoV infection. METHODS: The potential signaling pathways were manipulated individually by pharmacological inhibition, small interfering ribonucleic acid (siRNA) depletion, and antibody blocking. The MERS-CoV-induced proinflammatory response was evaluated by measuring the expression levels of key cytokines and/or chemokines. Reverse transcription-quantitative polymerase chain reaction assay, flow cytometry analysis, and Western blotting were applied to evaluate the activation of related PRRs and engagement of adaptors. RESULTS: MERS-CoV replication significantly upregulated C-type lectin receptor (CLR) macrophage-inducible Ca2+-dependent lectin receptor (Mincle). The role of Mincle for MERS-CoV-triggered cytokine/chemokine induction was established based on the results of antibody blockage, siRNA depletion of Mincle and its adaptor spleen tyrosine kinase (Syk), and Syk pharmacological inhibition. The cytokine and/or chemokine induction was significantly attenuated by siRNA depletion of retinoic acid-inducible-I-like receptors (RLR) or adaptor, indicating that RLR signaling also contributed to MERS-CoV-induced proinflammatory response. CONCLUSIONS: The CLR and RLR pathways are activated and contribute to the proinflammatory response in MERS-CoV-infected macrophages.


Assuntos
Infecções por Coronavirus/imunologia , Proteína DEAD-box 58/metabolismo , Lectinas Tipo C/metabolismo , Macrófagos/imunologia , Macrófagos/virologia , Coronavírus da Síndrome Respiratória do Oriente Médio/metabolismo , Animais , Proteínas Adaptadoras de Sinalização CARD , Quimiocinas/metabolismo , Chlorocebus aethiops , Infecções por Coronavirus/metabolismo , Infecções por Coronavirus/virologia , Citocinas/imunologia , Citocinas/metabolismo , Proteína DEAD-box 58/genética , Técnicas de Silenciamento de Genes , Humanos , Lectinas Tipo C/genética , Pulmão/imunologia , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , Coronavírus da Síndrome Respiratória do Oriente Médio/patogenicidade , RNA Interferente Pequeno/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Transdução de Sinais , Transcriptoma , Tretinoína/farmacologia , Células Vero , Replicação Viral
4.
J Virol ; 90(20): 9114-27, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27489282

RESUMO

UNLABELLED: The spike proteins of coronaviruses are capable of binding to a wide range of cellular targets, which contributes to the broad species tropism of coronaviruses. Previous reports have demonstrated that Middle East respiratory syndrome coronavirus (MERS-CoV) predominantly utilizes dipeptidyl peptidase 4 (DPP4) for cell entry. However, additional cellular binding targets of the MERS-CoV spike protein that may augment MERS-CoV infection have not been further explored. In the current study, using the virus overlay protein binding assay (VOPBA), we identified carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) as a novel cell surface binding target of MERS-CoV. CEACAM5 coimmunoprecipitated with the spike protein of MERS-CoV in both overexpressed and endogenous settings. Disrupting the interaction between CEACAM5 and MERS-CoV spike with anti-CEACAM5 antibody, recombinant CEACAM5 protein, or small interfering RNA (siRNA) knockdown of CEACAM5 significantly inhibited the entry of MERS-CoV. Recombinant expression of CEACAM5 did not render nonpermissive baby hamster kidney (BHK21) cells susceptible to MERS-CoV infection. Instead, CEACAM5 overexpression significantly enhanced the attachment of MERS-CoV to the BHK21 cells. More importantly, the entry of MERS-CoV was increased when CEACAM5 was overexpressed in permissive cells, which suggested that CEACAM5 could facilitate MERS-CoV entry in conjunction with DPP4 despite not being able to support MERS-CoV entry independently. Taken together, the results of our study identified CEACAM5 as a novel cell surface binding target of MERS-CoV that facilitates MERS-CoV infection by augmenting the attachment of the virus to the host cell surface. IMPORTANCE: Infection with the Middle East respiratory syndrome coronavirus (MERS-CoV) is associated with the highest mortality rate among all known human-pathogenic coronaviruses. Currently, there are no approved vaccines or therapeutics against MERS-CoV infection. The identification of carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) as a novel cell surface binding target of MERS-CoV advanced our knowledge on the cell binding biology of MERS-CoV. Importantly, CEACAM5 could potentiate the entry of MERS-CoV by functioning as an attachment factor. In this regard, CEACAM5 could serve as a novel target, in addition to dipeptidyl peptidase-4 (DPP4), in the development of antiviral strategies for MERS-CoV.


Assuntos
Antígeno Carcinoembrionário/metabolismo , Coronavírus da Síndrome Respiratória do Oriente Médio/fisiologia , Receptores Virais/metabolismo , Ligação Viral , Internalização do Vírus , Animais , Linhagem Celular , Proteínas Ligadas por GPI/metabolismo , Humanos
5.
J Infect Dis ; 213(6): 904-14, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26203058

RESUMO

Middle East respiratory syndrome (MERS) is associated with a mortality rate of >35%. We previously showed that MERS coronavirus (MERS-CoV) could infect human macrophages and dendritic cells and induce cytokine dysregulation. Here, we further investigated the interplay between human primary T cells and MERS-CoV in disease pathogenesis. Importantly, our results suggested that MERS-CoV efficiently infected T cells from the peripheral blood and from human lymphoid organs, including the spleen and the tonsil. We further demonstrated that MERS-CoV infection induced apoptosis in T cells, which involved the activation of both the extrinsic and intrinsic apoptosis pathways. Remarkably, immunostaining of spleen sections from MERS-CoV-infected common marmosets demonstrated the presence of viral nucleoprotein in their CD3(+) T cells. Overall, our results suggested that the unusual capacity of MERS-CoV to infect T cells and induce apoptosis might partly contribute to the high pathogenicity of the virus.


Assuntos
Apoptose/fisiologia , Coronavírus da Síndrome Respiratória do Oriente Médio/fisiologia , Linfócitos T/virologia , Animais , Anticorpos Antivirais , Callithrix , Células Cultivadas , Dipeptidil Peptidase 4/genética , Dipeptidil Peptidase 4/metabolismo , Regulação da Expressão Gênica , Humanos , Tonsila Palatina/citologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , Baço/citologia , Linfócitos T/fisiologia
6.
J Infect Dis ; 209(9): 1331-42, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24065148

RESUMO

Middle East respiratory syndrome coronavirus (MERS-CoV) infection caused severe pneumonia and multiorgan dysfunction and had a higher crude fatality rate (around 50% vs. 10%) than SARS coronavirus (SARS-CoV) infection. To understand the pathogenesis, we studied viral replication, cytokine/chemokine response, and antigen presentation in MERS-CoV-infected human monocyte-derived macrophages (MDMs) versus SARS-CoV-infected MDMs. Only MERS-CoV can replicate in MDMs. Both viruses were unable to significantly stimulate the expression of antiviral cytokines (interferon α [IFN-α] and IFN-ß) but induced comparable levels of tumor necrosis factor α and interleukin 6. Notably, MERS-CoV induced significantly higher expression levels of interleukin 12, IFN-γ, and chemokines (IP-10/CXCL-10, MCP-1/CCL-2, MIP-1α/CCL-3, RANTES/CCL-5, and interleukin 8) than SARS-CoV. The expression of major histocompatibility complex class I and costimulatory molecules were significantly higher in MERS-CoV-infected MDMs than in SARS-CoV-infected cells. MERS-CoV replication was validated by immunostaining of infected MDMs and ex vivo lung tissue. We conclusively showed that MERS-CoV can establish a productive infection in human macrophages. The aberrant induction of inflammatory cytokines/chemokines could be important in the disease pathogenesis.


Assuntos
Infecções por Coronavirus/virologia , Coronavirus/fisiologia , Citocinas/imunologia , Macrófagos/virologia , Pneumonia Viral/virologia , Replicação Viral/fisiologia , Animais , Apresentação de Antígeno , Sobrevivência Celular/imunologia , Células Cultivadas , Chlorocebus aethiops , Coronavirus/imunologia , Coronavirus/patogenicidade , Infecções por Coronavirus/imunologia , Citocinas/biossíntese , Humanos , Pulmão/virologia , Macrófagos/imunologia , Pneumonia Viral/imunologia , Células Vero
7.
EBioMedicine ; 89: 104485, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36857860

RESUMO

BACKGROUND: Obesity is a worldwide epidemic and is considered a risk factor of severe manifestation of Coronavirus Disease 2019 (COVID-19). The pathogenicity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and host responses to infection, re-infection, and vaccination in individuals with obesity remain incompletely understood. METHODS: Using the diet-induced obese (DIO) mouse model, we studied SARS-CoV-2 Alpha- and Omicron BA.1-induced disease manifestations and host immune responses to infection, re-infection, and COVID-19 mRNA vaccination. FINDINGS: Unlike in lean mice, Omicron BA.1 and Alpha replicated to comparable levels in the lungs of DIO mice and resulted in similar degree of tissue damages. Importantly, both T cell and B cell mediated adaptive immune responses to SARS-CoV-2 infection or COVID-19 mRNA vaccination are impaired in DIO mice, leading to higher propensity of re-infection and lower vaccine efficacy. However, despite the absence of neutralizing antibody, vaccinated DIO mice are protected from lung damage upon Omicron challenge, accompanied with significantly more IFN-α and IFN-ß production in the lung tissue. Lung RNAseq and subsequent experiments indicated that COVID-19 mRNA vaccination in DIO mice boosted antiviral innate immune response, including the expression of IFN-α, when compared to the nonvaccinated controls. INTERPRETATION: Our findings suggested that COVID-19 mRNA vaccination enhances host innate antiviral responses in obesity which protect the DIO mice to a certain degree when adaptive immunity is suboptimal. FUNDING: A full list of funding bodies that contributed to this study can be found in the Acknowledgements section.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Animais , Humanos , Camundongos , SARS-CoV-2 , Camundongos Obesos , Reinfecção , Dieta , Obesidade , Anticorpos Neutralizantes , Interferon-alfa , RNA Mensageiro , Antivirais , Anticorpos Antivirais , Vacinas de mRNA
8.
Emerg Microbes Infect ; 12(1): 2207678, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37122119

RESUMO

SUMMARY: Intranasal infection of newly-weaned Syrian hamsters by SARS-CoV-2 Omicron variants can lead to brain inflammation and neuron degeneration with detectable low level of viral load and sparse expression of viral nucleoprotein.


Assuntos
COVID-19 , Encefalite , Animais , Cricetinae , SARS-CoV-2 , Mesocricetus , Encéfalo
9.
Sci Adv ; 7(25)2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34134991

RESUMO

Infection by highly pathogenic coronaviruses results in substantial apoptosis. However, the physiological relevance of apoptosis in the pathogenesis of coronavirus infections is unknown. Here, with a combination of in vitro, ex vivo, and in vivo models, we demonstrated that protein kinase R-like endoplasmic reticulum kinase (PERK) signaling mediated the proapoptotic signals in Middle East respiratory syndrome coronavirus (MERS-CoV) infection, which converged in the intrinsic apoptosis pathway. Inhibiting PERK signaling or intrinsic apoptosis both alleviated MERS pathogenesis in vivo. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and SARS-CoV induced apoptosis through distinct mechanisms but inhibition of intrinsic apoptosis similarly limited SARS-CoV-2- and SARS-CoV-induced apoptosis in vitro and markedly ameliorated the lung damage of SARS-CoV-2-inoculated human angiotensin-converting enzyme 2 (hACE2) mice. Collectively, our study provides the first evidence that virus-induced apoptosis is an important disease determinant of highly pathogenic coronaviruses and demonstrates that this process can be targeted to attenuate disease severity.


Assuntos
Antivirais/farmacologia , Apoptose/efeitos dos fármacos , Tratamento Farmacológico da COVID-19 , Infecções por Coronavirus/tratamento farmacológico , eIF-2 Quinase/metabolismo , Adenina/análogos & derivados , Adenina/farmacologia , Enzima de Conversão de Angiotensina 2/genética , Animais , Apoptose/fisiologia , COVID-19/etiologia , COVID-19/patologia , Linhagem Celular , Infecções por Coronavirus/etiologia , Infecções por Coronavirus/patologia , Dipeptidil Peptidase 4/genética , Células Epiteliais/virologia , Feminino , Humanos , Indóis/farmacologia , Pulmão/virologia , Masculino , Camundongos Transgênicos , eIF-2 Quinase/antagonistas & inibidores , eIF-2 Quinase/genética
10.
EMBO Mol Med ; 11(1)2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30498026

RESUMO

Glycine decarboxylase (GLDC) was prioritized as a candidate susceptibility gene to severe influenza in humans. The higher expression of GLDC derived from genetic variations may confer a higher risk to H7N9 and severe H1N1 infection. We sought to characterize GLDC as functional susceptibility gene that GLDC may intrinsically regulate antiviral response, thereby impacting viral replication and disease outcome. We demonstrated that GLDC inhibitor AOAA and siRNA depletion boosted IFNß- and IFN-stimulated genes (ISGs) in combination with PolyI:C stimulation. GLDC inhibition and depletion significantly amplified antiviral response of type I IFNs and ISGs upon viral infection and suppressed the replication of H1N1 and H7N9 viruses. Consistently, GLDC overexpression significantly promoted viral replication due to the attenuated antiviral responses. Moreover, GLDC inhibition in H1N1-infected BALB/c mice recapitulated the amplified antiviral response and suppressed viral growth. AOAA provided potent protection to the infected mice from lethal infection, comparable to a standard antiviral against influenza viruses. Collectively, GLDC regulates cellular antiviral response and orchestrates viral growth. GLDC is a functional susceptibility gene to severe influenza in humans.


Assuntos
Predisposição Genética para Doença , Glicina Desidrogenase (Descarboxilante)/genética , Imunidade Inata , Influenza Humana/genética , Animais , Modelos Animais de Doenças , Inibidores Enzimáticos/administração & dosagem , Humanos , Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H1N1/imunologia , Subtipo H7N9 do Vírus da Influenza A/crescimento & desenvolvimento , Subtipo H7N9 do Vírus da Influenza A/imunologia , Influenza Humana/patologia , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções por Orthomyxoviridae/patologia , Tacrolimo/administração & dosagem , Tacrolimo/análogos & derivados , Resultado do Tratamento , Replicação Viral
11.
Emerg Microbes Infect ; 7(1): 47, 2018 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-29593225

RESUMO

To elucidate the adaptive strategies of influenza A viruses (IAVs) to human, we proposed a computational approach to identify human-adaptive mutations in seasonal IAVs, which have not been analyzed comprehensively. We compared representative PB2 sequences of 1425 avian IAVs and 2176 human IAVs and identified a total of 42 human-adaptive markers, including 28 and 31 markers in PB2 proteins of seasonal viruses H1N1 and H3N2, respectively. Notably, this comprehensive list encompasses almost all the markers identified in prior computational studies and 21 novel markers including an experimentally verified mutation K526R, suggesting the predictive power of our method. The strength of our analysis derives from the enormous amount of recently available sequences as well as the recognition that human-adaptive mutations are not necessarily conserved across subtypes. We also utilized mutual information to profile the inter-residue coevolution in PB2 protein. A total of 35 and 46 coevolving site pairs are identified in H1N1 and H3N2, respectively. Interestingly, 13 out of the 28 (46.4%) identified markers in H1N1 and 16 out of the 31 (51.6%) in H3N2 are embraced in the coevolving pairs. Many of them are paired with well-characterized human-adaptive mutations, indicating potential epistatic effect of these coevolving residues in human adaptation. Additionally, we reconstructed the PB2 evolutionary history of seasonal IAVs and demonstrated the distinct adaptive pathway of PB2 segment after reassortment from H1 to H3 lineage. Our study may provide clues for further experimental validation of human-adaptive mutations and shed light on the human adaptation process of seasonal IAVs.


Assuntos
Adaptação Biológica/genética , Vírus da Influenza A Subtipo H1N1/genética , Influenza Aviária/virologia , Influenza Humana/virologia , RNA Polimerase Dependente de RNA/genética , Análise de Sequência , Proteínas Virais/genética , Animais , Aves/virologia , Evolução Molecular , Genoma Viral , Humanos , Vírus da Influenza A Subtipo H1N1/classificação , Vírus da Influenza A Subtipo H3N2/genética , Vírus da Influenza A/genética , Influenza Aviária/transmissão , Influenza Humana/transmissão , Mutação , Vírus Reordenados/genética , Estações do Ano
12.
Sci Adv ; 3(11): eaao4966, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29152574

RESUMO

Middle East respiratory syndrome coronavirus (MERS-CoV) has caused human respiratory infections with a high case fatality rate since 2012. However, the mode of virus transmission is not well understood. The findings of epidemiological and virological studies prompted us to hypothesize that the human gastrointestinal tract could serve as an alternative route to acquire MERS-CoV infection. We demonstrated that human primary intestinal epithelial cells, small intestine explants, and intestinal organoids were highly susceptible to MERS-CoV and can sustain robust viral replication. We also identified the evidence of enteric MERS-CoV infection in the stool specimen of a clinical patient. MERS-CoV was considerably resistant to fed-state gastrointestinal fluids but less tolerant to highly acidic fasted-state gastric fluid. In polarized Caco-2 cells cultured in Transwell inserts, apical MERS-CoV inoculation was more effective in establishing infection than basolateral inoculation. Notably, direct intragastric inoculation of MERS-CoV caused a lethal infection in human DPP4 transgenic mice. Histological examination revealed MERS-CoV enteric infection in all inoculated mice, as shown by the presence of virus-positive cells, progressive inflammation, and epithelial degeneration in small intestines, which were exaggerated in the mice pretreated with the proton pump inhibitor pantoprazole. With the progression of the enteric infection, inflammation, virus-positive cells, and live viruses emerged in the lung tissues, indicating the development of sequential respiratory infection. Taken together, these data suggest that the human intestinal tract may serve as an alternative infection route for MERS-CoV.


Assuntos
Infecções por Coronavirus/patologia , Coronavírus da Síndrome Respiratória do Oriente Médio/fisiologia , Animais , Células CACO-2 , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/virologia , Dipeptidil Peptidase 4/genética , Dipeptidil Peptidase 4/metabolismo , Células Epiteliais/citologia , Células Epiteliais/virologia , Fezes/virologia , Feminino , Humanos , Intestino Delgado/citologia , Intestino Delgado/patologia , Intestino Delgado/virologia , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Transgênicos , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , Coronavírus da Síndrome Respiratória do Oriente Médio/patogenicidade , Pantoprazol/uso terapêutico , RNA Viral/genética , RNA Viral/metabolismo
13.
Virology ; 498: 1-8, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27525812

RESUMO

To evaluate the pathogenicity, a highly pathogenic avian influenza H7N7 virus (A/Netherlands/219/03) isolated from human was passaged in mice. A mutant virus (mH7N7) with attenuated virulence was isolated from mouse lung, which had a 3-log higher MLD50 than the wild-type virus (wH7N7). Sequence analysis and reverse genetics study revealed that mutations in PA account for the compromised viral replication in mammalian cells and mice. A mini-genome assay demonstrated that PA mutations P103H and S659L can cooperatively decrease polymerase activity. Actually, PA with double mutation P103H-S659L cannot sustain the generation of live virus by reverse genetics. Interestingly, the prior infection of mH7N7 virus provided mice with cross-protection against lethal challenge of other subtypes of influenza A virus including H1N1, H5N1 and H7N9. In conclusion, we demonstrated that PA mutations P103H and S659L can cooperatively reduce polymerase activity and viral replication in mammalian cells and attenuate pathogenicity in mice.


Assuntos
Substituição de Aminoácidos , Interações Hospedeiro-Patógeno , Vírus da Influenza A Subtipo H7N7/genética , Vírus da Influenza A Subtipo H7N7/patogenicidade , RNA Polimerase Dependente de RNA/genética , Proteínas Virais/genética , Animais , Linhagem Celular , Células Cultivadas , Feminino , Humanos , Camundongos , Mutação , Infecções por Orthomyxoviridae/mortalidade , Infecções por Orthomyxoviridae/virologia , Polimorfismo Genético , Carga Viral , Virulência/genética , Fatores de Virulência/genética , Replicação Viral
14.
Sci Rep ; 5: 8517, 2015 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-25687228

RESUMO

The fatality of avian influenza A(H7N9) infection in humans was over 30%. To identify human genetic susceptibility to A(H7N9) infection, we performed a genome-wide association study (GWAS) involving 102 A(H7N9) patients and 106 heavily-exposed healthy poultry workers, a sample size critically restricted by the small number of human A(H7N9) cases. To tackle the stringent significance cutoff of GWAS, we utilized an artificial imputation program SnipSnip to improve the association signals. In single-SNP analysis, one of the top SNPs was rs13057866 of LGALS1. The artificial imputation (AI) identified three non-genotyped causal variants, which can be represented by three anchor/partner SNP pairs rs13057866/rs9622682 (AI P = 1.81 × 10(-7)), rs4820294/rs2899292 (2.13 × 10(-7)) and rs62236673/rs2899292 (4.25 × 10(-7)) respectively. Haplotype analysis of rs4820294 and rs2899292 could simulate the signal of a causal variant. The rs4820294/rs2899292 haplotype GG, in association with protection from A(H7N9) infection (OR = 0.26, P = 5.92 × 10(-7)) correlated to significantly higher levels of LGALS1 mRNA (P = 0.050) and protein expression (P = 0.025) in lymphoblast cell lines. Additionally, rs4820294 was mapped as an eQTL in human primary monocytes and lung tissues. In conclusion, functional variants of LGALS1 causing the expression variations are contributable to the differential susceptibility to influenza A(H7N9).


Assuntos
Galectina 1/genética , Regulação da Expressão Gênica , Predisposição Genética para Doença , Subtipo H7N9 do Vírus da Influenza A , Influenza Humana/genética , Influenza Humana/virologia , Idoso , Linhagem Celular , Comorbidade , Fazendeiros , Feminino , Variação Genética , Estudo de Associação Genômica Ampla , Haplótipos , Humanos , Influenza Humana/epidemiologia , Influenza Humana/metabolismo , Desequilíbrio de Ligação , Masculino , Pessoa de Meia-Idade , Avaliação de Resultados da Assistência ao Paciente , Polimorfismo de Nucleotídeo Único , Reprodutibilidade dos Testes , Transdução de Sinais
15.
Virology ; 454-455: 197-205, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24725946

RESUMO

The Middle East respiratory syndrome coronavirus (MERS-CoV) closely resembled severe acute respiratory syndrome coronavirus (SARS-CoV) in disease manifestation as rapidly progressive acute pneumonia with multi-organ dysfunction. Using monocyte-derived-dendritic cells (Mo-DCs), we discovered fundamental discrepancies in the outcome of MERS-CoV- and SARS-CoV-infection. First, MERS-CoV productively infected Mo-DCs while SARS-CoV-infection was abortive. Second, MERS-CoV induced significantly higher levels of IFN-γ, IP-10, IL-12, and RANTES expression than SARS-CoV. Third, MERS-CoV-infection induced higher surface expression of MHC class II (HLA-DR) and the co-stimulatory molecule CD86 than SARS-CoV-infection. Overall, our data suggests that the dendritic cell can serve as an important target of viral replication and a vehicle for dissemination. MERS-CoV-infection in DCs results in the production of a rich combination of cytokines and chemokines, and modulates innate immune response differently from that of SARS-CoV-infection. Our findings may help to explain the apparent discrepancy in the pathogenicity between MERS-CoV and SARS-CoV.


Assuntos
Coronavirus/crescimento & desenvolvimento , Coronavirus/imunologia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Imunidade Inata , Antígeno B7-2/metabolismo , Células Cultivadas , Citocinas/metabolismo , Voluntários Saudáveis , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA