Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(17): e2300376120, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-37068258

RESUMO

The high transmissibility of SARS-CoV-2 Omicron subvariants was generally ascribed to immune escape. It remained unclear whether the emerging variants have gradually acquired replicative fitness in human respiratory epithelial cells. We sought to evaluate the replicative fitness of BA.5 and earlier variants in physiologically active respiratory organoids. BA.5 exhibited a dramatically increased replicative capacity and infectivity than B.1.1.529 and an ancestral strain wildtype (WT) in human nasal and airway organoids. BA.5 spike pseudovirus showed a significantly higher entry efficiency than that carrying WT or B.1.1.529 spike. Notably, we observed prominent syncytium formation in BA.5-infected nasal and airway organoids, albeit elusive in WT- and B.1.1.529-infected organoids. BA.5 spike-triggered syncytium formation was verified by lentiviral overexpression of spike in nasal organoids. Moreover, BA.5 replicated modestly in alveolar organoids, with a significantly lower titer than B.1.1.529 and WT. Collectively, the higher entry efficiency and fusogenic activity of BA.5 spike potentiated viral spread through syncytium formation in the human airway epithelium, leading to enhanced replicative fitness and immune evasion, whereas the attenuated replicative capacity of BA.5 in the alveolar organoids may account for its benign clinical manifestation.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , COVID-19/genética , Nariz , Organoides , Glicoproteína da Espícula de Coronavírus/genética , Anticorpos Neutralizantes , Anticorpos Antivirais
2.
J Med Virol ; 94(11): 5415-5424, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35791459

RESUMO

Human enterovirus A71 (EV-A71) is the major causative agent of hand-foot-and-mouth disease (HFMD) commonly associated with severe neurological diseases, particularly in children under 5 years of age. Several investigational therapeutic agents and vaccine candidates are being developed. However, no approved drug against EV-A71 infection is available, and no proven drug target has been identified. Since host kinases are key regulators of multiple signaling pathways in response to viral infections, here we screened a kinase inhibitor library and identified potent inhibitors against EV-A71 infection. Among the hits, GSK269962A, a Rho Associated Coiled-Coil Containing Protein Kinase (Rock) inhibitor with potent antiviral activity, was selected for further analysis. We found that this Rock inhibitor not only efficiently suppressed the replication of EV-A71 in RD cells, but also in human intestinal organoids, in a dose-dependent manner. Interestingly, small interfering RNA depletion of Rock1, but not Rock2, significantly restricted viral replication in RD cells, indicating that Rock1 is a novel host dependency factor for EV-A71 replication and can serve as a target for the development of anti-EV-A71 therapeutics.


Assuntos
Enterovirus Humano A , Infecções por Enterovirus , Enterovirus , Doença de Mão, Pé e Boca , Animais , Antígenos Virais , Criança , Pré-Escolar , Enterovirus Humano A/genética , Humanos , Quinases Associadas a rho
3.
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
4.
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
5.
Bio Protoc ; 13(8): e4657, 2023 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-37113328

RESUMO

A robust in vitro model of the human respiratory epithelium, including the alveolar and the airway epithelium, is essential for understanding the biology and pathology of the human respiratory system. We previously described a protocol to derive human lung organoids from primary lung tissues. We now describe a protocol to induce bidirectional differentiation to generate mature alveolar or airway organoids. The lung organoids are consecutively expanded for over one year with high stability, while the differentiated alveolar and airway organoids morphologically and functionally simulate the human alveolar and airway epithelium to a near-physiological level. Thus, we establish a robust organoid culture system of the entire human respiratory epithelium, the first two-phase bipotential organoid culture system that enables long-term expansion and bidirectional differentiation of respiratory epithelial cells. The long-term expandable lung organoids and differentiated organoids generate a stable and renewable source of respiratory epithelial cells, enabling scientists to reconstruct and expand the human respiratory epithelium in culture dishes. The respiratory organoid system provides a unique and physiologically active in vitro model of the human respiratory epithelium for various applications, including studying respiratory viral infection, disease modeling, drug screening, and pre-clinical testing. Graphical abstract.

6.
Viruses ; 15(5)2023 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-37243252

RESUMO

The respiratory epithelium, particularly the airway epithelium, is the primary infection site for respiratory pathogens. The apical surface of epithelial cells is constantly exposed to external stimuli including invading pathogens. Efforts have been made to establish organoid cultures to recapitulate the human respiratory tract. However, a robust and simple model with an easily accessible apical surface would benefit respiratory research. Here, we report the generation and characterization of apical-out airway organoids from the long-term expandable lung organoids that we previously established. The apical-out airway organoids morphologically and functionally recapitulated the human airway epithelium at a comparable level to the apical-in airway organoids. Moreover, apical-out airway organoids sustained productive and multicycle replication of SARS-CoV-2, and accurately recapitulated the higher infectivity and replicative fitness of the Omicron variants BA.5 and B.1.1.529 and an ancestral virus. In conclusion, we established a physiologically relevant and convenient apical-out airway organoid model for studying respiratory biology and diseases.


Assuntos
COVID-19 , Humanos , SARS-CoV-2 , Pulmão , Organoides
7.
Clin Exp Nephrol ; 16(6): 968-72, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23135867

RESUMO

Lupus vasculopathy (LV) is one of the complications of lupus nephritis. No definite therapy has been recommended for the management of LV, and patient outcomes are variable. We present the case of a 16-year-old girl who recovered from stage V chronic kidney disease due to lupus nephritis with vasculopathy with active treatment including pulse methylprednisolone, cyclophosphamide, rituximab and plasma exchange.


Assuntos
Nefrite Lúpica/complicações , Insuficiência Renal Crônica/etiologia , Insuficiência Renal Crônica/terapia , Índice de Gravidade de Doença , Doenças Vasculares/complicações , Adolescente , Anticorpos Monoclonais Murinos/uso terapêutico , Ciclofosfamida/uso terapêutico , Quimioterapia Combinada , Feminino , Humanos , Imunossupressores/uso terapêutico , Metilprednisolona/uso terapêutico , Troca Plasmática , Rituximab , Resultado do Tratamento , Doenças Vasculares/etiologia
8.
J Vis Exp ; (181)2022 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-35404361

RESUMO

The lack of a robust in vitro model of the human respiratory epithelium hinders the understanding of the biology and pathology of the respiratory system. We describe a defined protocol to derive human lung organoids from adult stem cells in the lung tissue and induce proximal differentiation to generate mature airway organoids. The lung organoids are then consecutively expanded for over 1 year with high stability, while the differentiated airway organoids are used to morphologically and functionally simulate human airway epithelium to a near-physiological level. Thus, we establish a robust organoid model of the human airway epithelium. The long-term expansion of lung organoids and differentiated airway organoids generates a stable and renewable source, enabling scientists to reconstruct and expand the human airway epithelial cells in culture dishes. The human lung organoid system provides a unique and physiologically active in vitro model for various applications, including studying virus-host interaction, drug testing, and disease modeling.


Assuntos
Pulmão , Organoides , Adulto , Diferenciação Celular , Células Epiteliais , Humanos , Tórax
9.
Signal Transduct Target Ther ; 7(1): 392, 2022 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-36529763

RESUMO

Horseshoe bats host numerous SARS-related coronaviruses without overt disease signs. Bat intestinal organoids, a unique model of bat intestinal epithelium, allow direct comparison with human intestinal organoids. We sought to unravel the cellular mechanism(s) underlying bat tolerance of coronaviruses by comparing the innate immunity in bat and human organoids. We optimized the culture medium, which enabled a consecutive passage of bat intestinal organoids for over one year. Basal expression levels of IFNs and IFN-stimulated genes were higher in bat organoids than in their human counterparts. Notably, bat organoids mounted a more rapid, robust and prolonged antiviral defense than human organoids upon Poly(I:C) stimulation. TLR3 and RLR might be the conserved pathways mediating antiviral response in bat and human intestinal organoids. The susceptibility of bat organoids to a bat coronavirus CoV-HKU4, but resistance to EV-71, an enterovirus of exclusive human origin, indicated that bat organoids adequately recapitulated the authentic susceptibility of bats to certain viruses. Importantly, TLR3/RLR inhibition in bat organoids significantly boosted viral growth in the early phase after SARS-CoV-2 or CoV-HKU4 infection. Collectively, the higher basal expression of antiviral genes, especially more rapid and robust induction of innate immune response, empowered bat cells to curtail virus propagation in the early phase of infection.


Assuntos
COVID-19 , Quirópteros , Viroses , Animais , Humanos , Quirópteros/genética , Antivirais/farmacologia , Receptor 3 Toll-Like/genética , SARS-CoV-2 , Organoides , Terapia de Imunossupressão
10.
mBio ; 13(4): e0194422, 2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-35938726

RESUMO

The human upper respiratory tract, specifically the nasopharyngeal epithelium, is the entry portal and primary infection site of respiratory viruses. Productive infection of SARS-CoV-2 in the nasal epithelium constitutes the cellular basis of viral pathogenesis and transmissibility. Yet a robust and well-characterized in vitro model of the nasal epithelium remained elusive. Here we report an organoid culture system of the nasal epithelium. We derived nasal organoids from easily accessible nasal epithelial cells with a perfect establishment rate. The derived nasal organoids were consecutively passaged for over 6 months. We then established differentiation protocols to generate 3-dimensional differentiated nasal organoids and organoid monolayers of 2-dimensional format that faithfully simulate the nasal epithelium. Moreover, when differentiated under a slightly acidic pH, the nasal organoid monolayers represented the optimal correlate of the native nasal epithelium for modeling the high infectivity of SARS-CoV-2, superior to all existing organoid models. Notably, the differentiated nasal organoid monolayers accurately recapitulated higher infectivity and replicative fitness of the Omicron variant than the prior variants. SARS-CoV-2, especially the more transmissible Delta and Omicron variants, destroyed ciliated cells and disassembled tight junctions, thereby facilitating virus spread and transmission. In conclusion, we establish a robust organoid culture system of the human nasal epithelium for modeling upper respiratory infections and provide a physiologically-relevant model for assessing the infectivity of SARS-CoV-2 emerging variants. IMPORTANCE An in vitro model of the nasal epithelium is imperative for understanding cell biology and virus-host interaction in the human upper respiratory tract. Here we report an organoid culture system of the nasal epithelium. Nasal organoids were derived from readily accessible nasal epithelial cells with perfect efficiency and stably expanded for more than 6 months. The long-term expandable nasal organoids were induced maturation into differentiated nasal organoids that morphologically and functionally simulate the nasal epithelium. The differentiated nasal organoids adequately recapitulated the higher infectivity and replicative fitness of SARS-CoV-2 emerging variants than the ancestral strain and revealed viral pathogenesis such as ciliary damage and tight junction disruption. Overall, we established a human nasal organoid culture system that enables a highly efficient reconstruction and stable expansion of the human nasal epithelium in culture plates, thus providing a facile and robust tool in the toolbox of microbiologists.


Assuntos
COVID-19 , Mucosa Nasal , Organoides , SARS-CoV-2 , COVID-19/virologia , Humanos , Mucosa Nasal/virologia , Organoides/virologia , SARS-CoV-2/classificação , SARS-CoV-2/patogenicidade , SARS-CoV-2/fisiologia , Técnicas de Cultura de Tecidos
11.
EMBO Mol Med ; 14(1): e14485, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-34826211

RESUMO

SPINK6 was identified in human skin as a cellular inhibitor of serine proteases of the KLK family. Airway serine proteases are required to cleave hemagglutinin (HA) of influenza A viruses (IAVs) to initiate an infection in the human airway. We hypothesized that SPINK6 may inhibit common airway serine proteases and restrict IAV activation. We demonstrate that SPINK6 specifically suppresses the proteolytic activity of HAT and KLK5, HAT- and KLK5-mediated HA cleavage, and restricts virus maturation and replication. SPINK6 constrains the activation of progeny virions and impairs viral growth; and vice versa, blocking endogenous SPINK6 enhances HA cleavage and viral growth in physiological-relevant human airway organoids where SPINK6 is intrinsically expressed. In IAV-infected mice, SPINK6 significantly suppresses viral growth and improves mouse survival. Notably, individuals carrying the higher SPINK6 expression allele were protected from human H7N9 infection. Collectively, SPINK6 is a novel host inhibitor of serine proteases in the human airway and restricts IAV activation.


Assuntos
Subtipo H7N9 do Vírus da Influenza A , Influenza Humana , Inibidores de Serinopeptidase do Tipo Kazal/metabolismo , Ativação Viral , Animais , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Humanos , Subtipo H7N9 do Vírus da Influenza A/fisiologia , Camundongos , Serina Proteases/metabolismo
12.
Cell Discov ; 8(1): 57, 2022 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-35710786

RESUMO

The airways and alveoli of the human respiratory tract are lined by two distinct types of epithelium, which are the primary targets of respiratory viruses. We previously established long-term expanding human lung epithelial organoids from lung tissues and developed a 'proximal' differentiation protocol to generate mucociliary airway organoids. However, a respiratory organoid system with bipotential of the airway and alveolar differentiation remains elusive. Here we defined a 'distal' differentiation approach to generate alveolar organoids from the same source for the derivation of airway organoids. The alveolar organoids consisting of type I and type II alveolar epithelial cells (AT1 and AT2, respectively) functionally simulate the alveolar epithelium. AT2 cells maintained in lung organoids serve as progenitor cells from which alveolar organoids derive. Moreover, alveolar organoids sustain a productive SARS-CoV-2 infection, albeit a lower replicative fitness was observed compared to that in airway organoids. We further optimized 2-dimensional (2D) airway organoids. Upon differentiation under a slightly acidic pH, the 2D airway organoids exhibit enhanced viral replication, representing an optimal in vitro correlate of respiratory epithelium for modeling the high infectivity of SARS-CoV-2. Notably, the higher infectivity and replicative fitness of the Omicron variant than an ancestral strain were accurately recapitulated in these optimized airway organoids. In conclusion, we have established a bipotential organoid culture system able to reproducibly expand the entire human respiratory epithelium in vitro for modeling respiratory diseases, including COVID-19.

13.
Stem Cell Reports ; 16(3): 493-504, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33626333

RESUMO

Enteroviruses, such as EV-A71 and CVA16, mainly infect the human gastrointestinal tract. Human coronaviruses, including SARS-CoV and SARS-CoV-2, have been variably associated with gastrointestinal symptoms. We aimed to optimize the human intestinal organoids and hypothesize that these optimized intestinal organoids can recapitulate enteric infections of enterovirus and coronavirus. We demonstrate that the optimized human intestinal organoids enable better simulation of the native human intestinal epithelium, and that they are significantly more susceptible to EV-A71 than CVA16. Higher replication of EV-A71 than CVA16 in the intestinal organoids triggers a more vigorous cellular response. However, SARS-CoV and SARS-CoV-2 exhibit distinct dynamics of virus-host interaction; more robust propagation of SARS-CoV triggers minimal cellular response, whereas, SARS-CoV-2 exhibits lower replication capacity but elicits a moderate cellular response. Taken together, the disparate profile of the virus-host interaction of enteroviruses and coronaviruses in human intestinal organoids may unravel the cellular basis of the distinct pathogenicity of these viral pathogens.


Assuntos
COVID-19/virologia , Enterovirus Humano A/patogenicidade , Infecções por Enterovirus/virologia , Intestinos/virologia , Organoides/virologia , SARS-CoV-2/patogenicidade , Animais , Linhagem Celular , Chlorocebus aethiops , Interações entre Hospedeiro e Microrganismos/fisiologia , Humanos , Mucosa Intestinal/virologia , Células Vero , Replicação Viral/fisiologia
14.
Clin Exp Nephrol ; 14(1): 75-9, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19802521

RESUMO

We report a 14-year-old girl with nephrotic syndrome and renal vein thrombosis (RVT) on initial presentation. The patient tested positive for antinuclear antibodies but only weakly positive for anti-double-stranded DNA (anti-dsDNA). Her C3 level was normal. Treatment with low molecular weight heparin resulted in resolution of RVT. Renal biopsy showed membranous glomerulonephritis with segmental sclerosis. Tissue immunostaining showed diffuse granular C3 and immunoglobulin (Ig)G staining along the capillary wall with focal segmental IgM staining deposits in the mesangium. No C1q, IgA, or fibrinogen was noted on immunofluorescence assay. With cyclosporin A and prednisolone, the patient went into remission and corticosteroids were tapered off gradually. Two years later, she had a relapse of proteinuria, hypocomplementemia, and extremely high anti-dsDNA. Systemic lupus erythematosus (SLE) was diagnosed, and she was promptly started on steroid and immunosuppressive agents, which resulted in reduction of proteinuria. Her renal function has been normal all along. Membranous nephropathy is uncommon in Chinese children and could be a possible early presentation of SLE.


Assuntos
Glomerulonefrite Membranosa/diagnóstico , Síndrome Nefrótica/complicações , Adolescente , Ciclosporina/uso terapêutico , DNA/imunologia , Enoxaparina/uso terapêutico , Feminino , Glomerulonefrite Membranosa/patologia , Humanos , Imunossupressores/uso terapêutico , Dor Lombar/diagnóstico , Lúpus Eritematoso Sistêmico/diagnóstico , Síndrome Nefrótica/diagnóstico , Prednisolona/uso terapêutico , Proteinúria/diagnóstico , Proteinúria/tratamento farmacológico , Proteinúria/etiologia , Veias Renais , Trombose Venosa/diagnóstico
15.
Emerg Microbes Infect ; 9(1): 2663-2672, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33179566

RESUMO

Rapid accumulation of viral proteins in host cells render viruses highly dependent on cellular chaperones including heat shock protein 90 (Hsp90). Three highly pathogenic human coronaviruses, including MERS-CoV, SARS-CoV and SARS-CoV-2, have emerged in the past 2 decades. However, there is no approved antiviral agent against these coronaviruses. We inspected the role of Hsp90 for coronavirus propagation. First, an Hsp90 inhibitor, 17-AAG, significantly suppressed MERS-CoV propagation in cell lines and physiological-relevant human intestinal organoids. Second, siRNA depletion of Hsp90ß, but not Hsp90α, significantly restricted MERS-CoV replication and abolished virus spread. Third, Hsp90ß interaction with MERS-CoV nucleoprotein (NP) was revealed in a co-immunoprecipitation assay. Hsp90ß is required to maintain NP stability. Fourth, 17-AAG substantially inhibited the propagation of SARS-CoV and SARS-CoV-2. Collectively, Hsp90 is a host dependency factor for human coronavirus MERS-CoV, SARS-CoV and SARS-COV-2. Hsp90 inhibitors can be repurposed as a potent and broad-spectrum antiviral against human coronaviruses.


Assuntos
Antivirais/farmacologia , Benzoquinonas/farmacologia , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/genética , Interações entre Hospedeiro e Microrganismos/efeitos dos fármacos , Lactamas Macrocíclicas/farmacologia , Coronavírus da Síndrome Respiratória do Oriente Médio/efeitos dos fármacos , Células A549 , Animais , Linhagem Celular , Chlorocebus aethiops , Células HEK293 , Humanos , Intestinos/virologia , Técnicas de Cultura de Órgãos , RNA Interferente Pequeno , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/efeitos dos fármacos , SARS-CoV-2/efeitos dos fármacos , Células Vero , Replicação Viral/efeitos dos fármacos , Tratamento Farmacológico da COVID-19
16.
Nat Med ; 26(7): 1077-1083, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32405028

RESUMO

A novel coronavirus-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-emerged in humans in Wuhan, China, in December 2019 and has since disseminated globally1,2. As of April 16, 2020, the confirmed case count of coronavirus disease 2019 (COVID-19) had surpassed 2 million. Based on full-genome sequence analysis, SARS-CoV-2 shows high homology to SARS-related coronaviruses identified in horseshoe bats1,2. Here we show the establishment and characterization of expandable intestinal organoids derived from horseshoe bats of the Rhinolophus sinicus species that can recapitulate bat intestinal epithelium. These bat enteroids are fully susceptible to SARS-CoV-2 infection and sustain robust viral replication. Development of gastrointestinal symptoms in some patients with COVID-19 and detection of viral RNA in fecal specimens suggest that SARS-CoV-2 might cause enteric, in addition to respiratory, infection3,4. Here we demonstrate active replication of SARS-CoV-2 in human intestinal organoids and isolation of infectious virus from the stool specimen of a patient with diarrheal COVID-19. Collectively, we established the first expandable organoid culture system of bat intestinal epithelium and present evidence that SARS-CoV-2 can infect bat intestinal cells. The robust SARS-CoV-2 replication in human intestinal organoids suggests that the human intestinal tract might be a transmission route of SARS-CoV-2.


Assuntos
Betacoronavirus/patogenicidade , Infecções por Coronavirus/patologia , Infecções por Coronavirus/transmissão , Intestinos/virologia , Organoides/virologia , Pneumonia Viral/patologia , Pneumonia Viral/transmissão , Animais , COVID-19 , Diferenciação Celular , Células Cultivadas , Pré-Escolar , Quirópteros/virologia , Chlorocebus aethiops , Infecções por Coronavirus/virologia , Enterócitos/patologia , Enterócitos/fisiologia , Enterócitos/virologia , Feminino , Humanos , Lactente , Mucosa Intestinal/patologia , Mucosa Intestinal/virologia , Intestinos/patologia , Masculino , Organoides/patologia , Pandemias , Pneumonia Viral/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , SARS-CoV-2 , Células Vero , Carga Viral/genética , Carga Viral/métodos , Tropismo Viral/fisiologia
17.
Pediatr Nephrol ; 24(9): 1699-703, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19444481

RESUMO

This retrospective study aimed to evaluate the applicability of the selective approach of imaging infants < 6 months old with urinary tract infection (UTI) according to the UTI guidelines of the National Institute for Health and Clinical Excellence (NICE) 2007. Infants < 6 months old with their first UTI from January 2001 to December 2006 having undergone an ultrasound examination of the urinary tract, a micturating cystourethrogram, and a late di-mercaptosuccinic acid (DMSA) scan, were included. Their condition was evaluated against a set of risk features according to the UTI guidelines. Those having any one of these were classified as atypical and those having none as typical. There were 134 infants reviewed, with a typical (98 infants) to atypical (36 infants) ratio of 2.7 to 1. Girls were found to be relatively more represented in the atypical group [male (M):female (F) = 1.3:1] than in the typical group (M:F = 4.4:1) (P < 0.004). There were significantly more infants with abnormal micturating voiding cystourethrograms (MCUGs) (P = 0.007), more refluxing ureters (P < 0.001) and more significant vesico-ureteral reflux (VUR) (>/= grade III) (P = 0.013) in the atypical group than in the typical group; while there was no significant difference in ultrasound (US) and DMSA scan findings between the two groups. In the atypical group there was no difference in imaging studies (and, thus, the results) between the conventional practice and the NICE UTI recommendation. In the typical group, if the recommendations of the guidelines had been followed (i.e. only those with abnormal US would have been further investigated), 25 refluxing ureters and 22 scarred kidneys would have been left undiagnosed. In conclusion, application of the suggested selective imaging approach would leave a significant number of VUR and renal scars undiagnosed, and it may not be an optimal practice for infants less than 6 months old with their first UTI. The best approach remains to be clarified.


Assuntos
Diagnóstico por Imagem/métodos , Infecções Urinárias/diagnóstico , Diagnóstico por Imagem/normas , Feminino , Humanos , Lactente , Masculino , Guias de Prática Clínica como Assunto , Valor Preditivo dos Testes , Compostos Radiofarmacêuticos , Reprodutibilidade dos Testes , Estudos Retrospectivos , Ácido Dimercaptossuccínico Tecnécio Tc 99m , Ultrassonografia , Infecções Urinárias/diagnóstico por imagem , Refluxo Vesicoureteral/diagnóstico , Refluxo Vesicoureteral/fisiopatologia
18.
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
19.
Antiviral Res ; 161: 125-133, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30503888

RESUMO

Human respiratory syncytial virus (HRSV) infection is a significant cause of morbidity and mortality, particularly among the children and the elderly. Despite extensive efforts, there is currently no formally approved vaccine and effective antiviral options against HRSV infection are limited. The development of vaccines and antiviral strategies for HRSV was partly hampered by the lack of an efficient lethal mouse model to evaluate the efficacy of the candidate vaccines or antivirals. In this study, we established a lethal HRSV mouse model by consecutively passaging a clinical HRSV isolate, GZ08-0. GZ08-18 was isolated from mouse bronchioalveolar lavage fluids at the 50th passage of GZ08-0. Importantly, all GZ08-18-inoculated mice succumbed to the infection by day 7 post infection, whereas all GZ08-0-inoculated mice recovered from the infection. Subsequent investigations demonstrated that GZ08-18 replicated to a higher titer in mouse lungs, induced more prominent lung pathology, and resulted in higher expression levels of a number of key pro-inflammatory cytokines including IFN-γ, MIP-1α, and TNF-α in comparison to that of GZ08-0. The cyclophosphamide pretreatment rendered the mice more susceptible to a lethal outcome with less rounds of virus inoculation. Full genome sequencing revealed 17 mutations in GZ08-18, some of which might account for the dramatically increased pathogenicity over GZ08-0. In addition, by using ribavirin as a positive control, we demonstrated the potential application of this lethal mouse model as a tool in HRSV investigations. Overall, we have successfully established a practical lethal mouse model for HRSV with a mouse-adapted virus, which may facilitate future in vivo studies on the evaluation of candidate vaccines and drugs against HRSV.


Assuntos
Envelhecimento , Modelos Animais de Doenças , Infecções por Vírus Respiratório Sincicial/mortalidade , Vírus Sincicial Respiratório Humano/patogenicidade , Animais , Antivirais/uso terapêutico , Linhagem Celular , Ciclofosfamida/farmacologia , Genoma Viral , Humanos , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Vírus Sincicial Respiratório Humano/fisiologia , Ribavirina/uso terapêutico , Replicação Viral
20.
Nat Commun ; 10(1): 120, 2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30631056

RESUMO

Viruses are obligate intracellular microbes that exploit the host metabolic machineries to meet their biosynthetic demands, making these host pathways potential therapeutic targets. Here, by exploring a lipid library, we show that AM580, a retinoid derivative and RAR-α agonist, is highly potent in interrupting the life cycle of diverse viruses including Middle East respiratory syndrome coronavirus and influenza A virus. Using click chemistry, the overexpressed sterol regulatory element binding protein (SREBP) is shown to interact with AM580, which accounts for its broad-spectrum antiviral activity. Mechanistic studies pinpoint multiple SREBP proteolytic processes and SREBP-regulated lipid biosynthesis pathways, including the downstream viral protein palmitoylation and double-membrane vesicles formation, that are indispensable for virus replication. Collectively, our study identifies a basic lipogenic transactivation event with broad relevance to human viral infections and represents SREBP as a potential target for the development of broad-spectrum antiviral strategies.


Assuntos
Benzoatos/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Proteínas de Ligação a Elemento Regulador de Esterol/metabolismo , Tetra-Hidronaftalenos/farmacologia , Replicação Viral/efeitos dos fármacos , Antivirais/farmacologia , Benzoatos/química , Benzoatos/metabolismo , Vias Biossintéticas/efeitos dos fármacos , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/fisiologia , Lipídeos/biossíntese , Coronavírus da Síndrome Respiratória do Oriente Médio/efeitos dos fármacos , Coronavírus da Síndrome Respiratória do Oriente Médio/fisiologia , Ligação Proteica , Retinoides/química , Retinoides/metabolismo , Retinoides/farmacologia , Tetra-Hidronaftalenos/química , Tetra-Hidronaftalenos/metabolismo , Viroses/prevenção & controle , Viroses/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA