Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 500
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 132(3): 449-62, 2008 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-18267075

RESUMO

Plant innate immunity relies on the recognition of pathogen effector molecules by nucleotide-binding-leucine-rich repeat (NB-LRR) immune receptor families. Previously we have shown the N immune receptor, a member of TIR-NB-LRR family, indirectly recognizes the 50 kDa helicase (p50) domain of Tobacco mosaic virus (TMV) through its TIR domain. We have identified an N receptor-interacting protein, NRIP1, that directly interacts with both N's TIR domain and p50. NRIP1 is a functional rhodanese sulfurtransferase and is required for N to provide complete resistance to TMV. Interestingly, NRIP1 that normally localizes to the chloroplasts is recruited to the cytoplasm and nucleus by the p50 effector. As a consequence, NRIP1 interacts with N only in the presence of the p50 effector. Our findings show that a chloroplastic protein is intimately involved in pathogen recognition. We propose that N's activation requires a prerecognition complex containing the p50 effector and NRIP1.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Nicotiana/imunologia , Proteínas Nucleares/imunologia , Doenças das Plantas/imunologia , Proteínas de Plantas/imunologia , Receptores Virais/imunologia , Vírus do Mosaico do Tabaco/imunologia , Proteínas Adaptadoras de Transdução de Sinal/análise , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Antígenos Virais/imunologia , Núcleo Celular/química , Cloroplastos/química , Citoplasma/química , Imunidade Inata , Proteínas Nucleares/análise , Proteínas Nucleares/metabolismo , Proteína 1 de Interação com Receptor Nuclear , Proteínas de Plantas/análise , Proteínas de Plantas/metabolismo , Estrutura Terciária de Proteína , Receptores Virais/análise , Receptores Virais/metabolismo , Tiossulfato Sulfurtransferase/metabolismo , Nicotiana/virologia , Técnicas do Sistema de Duplo-Híbrido
2.
J Cell Physiol ; 236(4): 2950-2958, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32944935

RESUMO

Coronavirus disease-2019 (COVID-19) is a global pandemic and caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which has resulted in millions of deaths worldwide. Reports denote SARS-CoV-2 uses angiotensin-converting enzyme 2 (ACE2), transmembrane serine protease 2 (TMPRSS2) as its primary entry point into the host cell. However, understanding the biology behind this viral replication, disease mechanism and drug discovery efforts are limited due to the lack of a suitable experimental model. Here, we used single-cell RNA sequencing data of human organoids to analyze expressions of ACE2 and TMPRSS2, in addition to an array of RNA receptors to examine their role in SARS-CoV-2 pathogenesis. ACE2 is abundant in all organoids, except the prostate and brain, and TMPRSS2 is omnipresent. Innate immune pathways are upregulated in ACE2(+) cells of all organoids, except the lungs. Besides this, the expression of low-density lipoprotein receptor is highly enriched in ACE2(+) cells in intestinal, lung, and retinal organoids, with the highest expression in lung organoids. Collectively, this study demonstrates that the organoids can be used as an experimental platform to explore this novel virus disease mechanism and for drug development.


Assuntos
Enzima de Conversão de Angiotensina 2/análise , COVID-19 , Organoides , Análise de Sequência de RNA/métodos , Serina Endopeptidases/análise , Análise de Célula Única/métodos , Humanos , Modelos Biológicos , Receptores Virais/análise , SARS-CoV-2 , Internalização do Vírus
3.
J Endocrinol Invest ; 44(5): 1085-1090, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33025553

RESUMO

PURPOSE: SARS-COV-2 is a pathogenic agent belonging to the coronavirus family, responsible for the current global world pandemic. Angiotensin-converting enzyme 2 (ACE-2) is the receptor for cellular entry of SARS-CoV-2. ACE-2 is a type I transmembrane metallo-carboxypeptidase involved in the Renin-Angiotensin pathway. By analyzing two independent databases, ACE-2 was identified in several human tissues including the thyroid. Although some cases of COVID-19-related subacute thyroiditis were recently described, direct proof for the expression of the ACE-2 mRNA in thyroid cells is still lacking. Aim of the present study was to investigate by RT-PCR whether the mRNA encoding for ACE-2 is present in human thyroid cells. METHODS: RT-PCR was performed on in vitro ex vivo study on thyroid tissue samples (15 patients undergoing thyroidectomy for benign thyroid nodules) and primary thyroid cell cultures. RESULTS: The ACE-2 mRNA was detected in all surgical thyroid tissue samples (n = 15). Compared with two reporter genes (GAPDH: 0.052 ± 0.0026 Cycles-1; ß-actin: 0.044 ± 0.0025 Cycles-1; ACE-2: 0.035 ± 0.0024 Cycles-1), the mean level of transcript expression for ACE-2 mRNA was abundant. The expression of ACE-2 mRNA in follicular cells was confirmed by analyzing primary cultures of thyroid cells, which expressed the ACE-2 mRNA at levels similar to tissues. CONCLUSIONS: The results of the present study demonstrate that the mRNA encoding for the ACE-2 receptor is expressed in thyroid follicular cells, making them a potential target for SARS-COV-2 entry. Future clinical studies in patients with COVID-19 will be required for increase our understanding of the thyroid repercussions of SARS-CoV-2 infection.


Assuntos
Enzima de Conversão de Angiotensina 2/análise , COVID-19/complicações , RNA Mensageiro/análise , Receptores Virais/análise , Tireoidite Subaguda/etiologia , Adulto , COVID-19/metabolismo , Feminino , Humanos , Masculino , Cultura Primária de Células , Reação em Cadeia da Polimerase em Tempo Real , Glândula Tireoide/química , Glândula Tireoide/citologia , Tireoidectomia , Tireoidite Subaguda/metabolismo
4.
Clin Gastroenterol Hepatol ; 18(9): 2128-2130.e2, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32334082

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a novel coronavirus that causes coronavirus disease 2019 (COVID-19) in human beings, has caused a serious public health issue.1 Attention to pancreatic injury is lacking, which may impact patients' prognosis. In this study, we explored the expression and distribution of angiotensin-converting enzyme 2 (ACE2), the receptor of SARS-CoV-2, in the pancreas. Combined with clinical data, we showed that pancreatic injury can occur in some COVID-19 patients.


Assuntos
Betacoronavirus/crescimento & desenvolvimento , Infecções por Coronavirus/complicações , Perfilação da Expressão Gênica , Pâncreas/enzimologia , Pancreatopatias/fisiopatologia , Peptidil Dipeptidase A/análise , Pneumonia Viral/complicações , Receptores Virais/análise , Adolescente , Adulto , Idoso , Enzima de Conversão de Angiotensina 2 , COVID-19 , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Pandemias , SARS-CoV-2 , Adulto Jovem
5.
Pancreatology ; 20(6): 1218-1225, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32828686

RESUMO

BACKGROUND: Among the several new targets for the comprehension of the biology of pancreatic ductal adenocarcinoma (PDAC), Prion proteins (PrPc) deserve particular mention, since they share a marked neurotropism. Actually, PrPc could have also a role in tumorigenesis, as recently demonstrated. However, only few in vitro studies in cell cultures showed the occurrence of PrPc in PDAC cells. We aim to evaluate the presence of PrPc in vivo in PDAC tissues as a potential new biomarker. METHODS: Samples from tumors of 23 patients undergone pancreatic resections from July 2018 to May 2020 at our institution were collected and analyzed. Immunohistochemistry and western blotting of PDAC tissues were compared with control tissues. Immunohistochemistry was used also to evaluate the localization of PrPc and of CD155, a tumoral stem-cell marker. RESULTS: All cases were moderately differentiated PDAC, with perineural invasion (PNI) in 19/23 cases (83%). According to western-blot analysis, PrPc was markedly expressed in PDAC tissues (273.5 ± 44.63 OD) respect to controls (100 ± 28.35 OD, p = 0.0018). Immunohistochemistry confirmed these findings, with higher linear staining of PrPc in PDAC ducts (127.145 ± 7.56 µm vs 75.21 ± 5.01 µm, p < 0.0001). PrPc and CD155 exactly overlapped in ductal tumoral cells, highlighting the possible relationship of PrPc with cancer stemness. Finally, PrPc expression related with cancer stage and there was a potential correspondence with PNI. CONCLUSIONS: Our work provides evidence for increased levels of PrPc in PDAC. This might contribute to cancer aggressiveness and provides a potentially new biomarker. Work is in progress to decipher clinical implications.


Assuntos
Adenocarcinoma/química , Adenocarcinoma/cirurgia , Neoplasias Pancreáticas/química , Neoplasias Pancreáticas/cirurgia , Proteínas Priônicas/química , Adenocarcinoma/patologia , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/análise , Western Blotting , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Células-Tronco Neoplásicas , Pancreatectomia , Neoplasias Pancreáticas/patologia , Proteínas Priônicas/genética , Prognóstico , Receptores Virais/análise
6.
Gastroenterol Hepatol ; 43(8): 464-471, 2020 Oct.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-32859408

RESUMO

The SARS-CoV-2 pandemic is leading to high mortality and a global health crisis. The primary involvement is respiratory; however, the virus can also affect other organs, such as the gastrointestinal tract and liver. The most common symptoms are anorexia and diarrhea. In about half of the cases, viral RNA could be detected in the stool, which is another line of transmission and diagnosis. covid19 has a worse prognosis in patients with comorbidities, although there is not enough evidence in case of previous digestive diseases. Digestive endoscopies may give rise to aerosols, which make them techniques with a high risk of infection. Experts and scientific organizations worldwide have developed guidelines for preventive measures. The available evidence on gastrointestinal and hepatic involvement, the impact on patients with previous digestive diseases and operating guidelines for Endoscopy Units during the pandemic are reviewed.


Assuntos
Betacoronavirus/patogenicidade , Infecções por Coronavirus/complicações , Doenças do Sistema Digestório/etiologia , Sistema Digestório/virologia , Pandemias , Pneumonia Viral/complicações , Aerossóis , Enzima de Conversão de Angiotensina 2 , Anorexia/etiologia , Antivirais/efeitos adversos , Betacoronavirus/isolamento & purificação , Betacoronavirus/fisiologia , COVID-19 , Estudos de Coortes , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/transmissão , Diarreia/etiologia , Doenças do Sistema Digestório/virologia , Endoscopia do Sistema Digestório/efeitos adversos , Fezes/virologia , Humanos , Imunossupressores/efeitos adversos , Intestinos/química , Intestinos/virologia , Hepatopatias/etiologia , Estudos Multicêntricos como Assunto , Pandemias/prevenção & controle , Peptidil Dipeptidase A/análise , Peptidil Dipeptidase A/fisiologia , Equipamento de Proteção Individual , Pneumonia Viral/tratamento farmacológico , Pneumonia Viral/prevenção & controle , Pneumonia Viral/transmissão , Receptores Virais/análise , Receptores Virais/fisiologia , Risco , SARS-CoV-2 , Precauções Universais , Tratamento Farmacológico da COVID-19
7.
Clin Infect Dis ; 66(1): 45-53, 2018 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-29020176

RESUMO

Background: Middle East respiratory syndrome coronavirus (MERS-CoV) causes pneumonia with a relatively high case fatality rate in humans. Smokers and chronic obstructive pulmonary disease (COPD) patients have been reported to be more susceptible to MERS-CoV infection. Here, we determined the expression of MERS-CoV receptor, dipeptidyl peptidase IV (DPP4), in lung tissues of smokers without airflow limitation and COPD patients in comparison to nonsmoking individuals (never-smokers). Methods: DPP4 expression was measured in lung tissue of lung resection specimens of never-smokers, smokers without airflow limitation, COPD GOLD stage II patients and in lung explants of end-stage COPD patients. Both control subjects and COPD patients were well phenotyped and age-matched. The mRNA expression was determined using qRT-PCR and protein expression was quantified using immunohistochemistry. Results: In smokers and subjects with COPD, both DPP4 mRNA and protein expression were significantly higher compared to never-smokers. Additionally, we found that both DPP4 mRNA and protein expression were inversely correlated with lung function and diffusing capacity parameters. Conclusions: We provide evidence that DPP4 is upregulated in the lungs of smokers and COPD patients, which could partially explain why these individuals are more susceptible to MERS-CoV infection. These data also highlight a possible role of DPP4 in COPD pathogenesis.


Assuntos
Dipeptidil Peptidase 4/análise , Pulmão/patologia , Doença Pulmonar Obstrutiva Crônica/patologia , Receptores Virais/análise , Fumar/efeitos adversos , Regulação para Cima , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Inquéritos e Questionários
8.
J Virol ; 90(3): 1455-69, 2016 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-26581996

RESUMO

UNLABELLED: H4 avian influenza virus (AIV) is one of the most prevalent influenza virus subtypes in the world. However, whether H4 AIVs pose a threat to public health remains largely unclear. Here, we analyzed the phylogenetic relationships, receptor binding properties, replication, and transmissibility in mammals of H4 AIVs isolated from live poultry markets in China between 2009 and 2012. Genomic sequence analysis of 36 representative H4 viruses revealed 32 different genotypes, indicating that these viruses are undergoing complex and frequent reassortment events. All 32 viruses tested could replicate in the respiratory organs of infected mice without prior adaptation. Receptor binding analysis demonstrated that the H4 AIVs bound to α-2,6-linked glycans, although they retained the binding preference for α-2,3-linked glycans. When we tested the direct-contact transmission of 10 H4 viruses in guinea pigs, we found that three viruses did not transmit to any of the contact animals, one virus transmitted to one of three contact animals, and six viruses transmitted to all three contact animals. When we further tested the respiratory droplet transmissibility of four of the viruses that transmitted efficiently via direct contact, we found that three of them could transmit to one or two of the five exposed animals. Our study demonstrates that the current circulating H4 AIVs can infect, replicate in, and transmit to mammalian hosts, thereby posing a potential threat to human health. These findings emphasize the continual need for enhanced surveillance of H4 AIVs. IMPORTANCE: Numerous surveillance studies have documented the wide distribution of H4 AIVs throughout the world, yet the biological properties of H4 viruses have not been well studied. In this study, we found that multiple genotypes of H4 viruses are cocirculating in the live poultry markets of China and that H4 viruses can replicate in mice, possess human-type receptor binding specificity, and transmit between guinea pigs via direct contact. Strikingly, some H4 strains also can transmit via respiratory droplet, albeit with limited efficiency. These results clearly show the potential threat posed by H4 viruses to public health.


Assuntos
Vírus da Influenza A/crescimento & desenvolvimento , Vírus da Influenza A/isolamento & purificação , Influenza Aviária/virologia , Infecções por Orthomyxoviridae/veterinária , Aves Domésticas/virologia , Ligação Viral , Replicação Viral , Animais , China , Análise por Conglomerados , Feminino , Genoma Viral , Cobaias , Vírus da Influenza A/genética , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Infecções por Orthomyxoviridae/transmissão , Filogenia , RNA Viral/genética , Receptores Virais/análise , Análise de Sequência de DNA , Homologia de Sequência
9.
Cell Tissue Res ; 370(2): 267-273, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28766044

RESUMO

Primary familial brain calcification (PFBC) is a neuropsychiatric disorder characterized by bilateral cerebral calcification with diverse neurologic or psychiatric symptoms. Recently, XPR1 variation has accounted for PFBC as another new causative gene. However, little is known about the distribution and basic function of XPR1 and its interaction with the other three pathogenic genes for PFBC (SLC20A2, PDGFRB and PDGFB). The aim of this study was to further clarify the role of XPR1 in PFBC brain pathology. As a result, gene expression profiles showed that XPR1 mRNA was widely expressed throughout the mouse brain. Cerebellum and striatum, most commonly affected in PFBC, contained a higher level of XPR1 protein than other brain regions. Additionally, XPR1 deficiency seriously affected Pi efflux and XPR1 mutations seemed to have an effect through haploinsufficiency mechanism. The immunoprecipitation and immunohistochemical studies demonstrated that XPR1 could interact with PDGFRB and might form a complex on the cell membrane. These results suggested that XPR1 played a fundamental role in the maintenance of cellular phosphate balance in the brain. This provided us with a novel perspective on understanding the pathophysiology of PFBC. The expression networks and interaction with the known pathogenic genes could shed new light on additional candidate genes for PFBC.


Assuntos
Encefalopatias/genética , Encéfalo/metabolismo , Calcinose/genética , Receptores Acoplados a Proteínas G/genética , Receptores Virais/genética , Transcriptoma , Animais , Encéfalo/patologia , Encefalopatias/metabolismo , Encefalopatias/patologia , Calcinose/metabolismo , Calcinose/patologia , Expressão Gênica , Predisposição Genética para Doença , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mapas de Interação de Proteínas , RNA Mensageiro/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/análise , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores Acoplados a Proteínas G/análise , Receptores Acoplados a Proteínas G/metabolismo , Receptores Virais/análise , Receptores Virais/metabolismo , Regulação para Cima , Receptor do Retrovírus Politrópico e Xenotrópico
10.
J Virol ; 89(14): 7202-13, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25926653

RESUMO

UNLABELLED: Human coronavirus (hCoV) HKU1 is one of six hCoVs identified to date and the only one with an unidentified cellular receptor. hCoV-HKU1 encodes a hemagglutinin-esterase (HE) protein that is unique to the group a betacoronaviruses (group 2a). The function of HKU1-HE remains largely undetermined. In this study, we examined binding of the S1 domain of hCoV-HKU1 spike to a panel of cells and found that the S1 could specifically bind on the cell surface of a human rhabdomyosarcoma cell line, RD. Pretreatment of RD cells with neuraminidase (NA) and trypsin greatly reduced the binding, suggesting that the binding was mediated by sialic acids on glycoproteins. However, unlike other group 2a CoVs, e.g., hCoV-OC43, for which 9-O-acetylated sialic acid (9-O-Ac-Sia) serves as a receptor determinant, HKU1-S1 bound with neither 9-O-Ac-Sia-containing glycoprotein(s) nor rat and mouse erythrocytes. Nonetheless, the HKU1-HE was similar to OC43-HE, also possessed sialate-O-acetylesterase activity, and acted as a receptor-destroying enzyme (RDE) capable of eliminating the binding of HKU1-S1 to RD cells, whereas the O-acetylesterase-inactive HKU1-HE mutant lost this capacity. Using primary human ciliated airway epithelial (HAE) cell cultures, the only in vitro replication model for hCoV-HKU1 infection, we confirmed that pretreatment of HAE cells with HE but not the enzymatically inactive mutant blocked hCoV-HKU1 infection. These results demonstrate that hCoV-HKU1 exploits O-Ac-Sia as a cellular attachment receptor determinant to initiate the infection of host cells and that its HE protein possesses the corresponding sialate-O-acetylesterase RDE activity. IMPORTANCE: Human coronaviruses (hCoV) are important human respiratory pathogens. Among the six hCoVs identified to date, only hCoV-HKU1 has no defined cellular receptor. It is also unclear whether hemagglutinin-esterase (HE) protein plays a role in viral entry. In this study, we found that, similarly to other members of the group 2a CoVs, sialic acid moieties on glycoproteins are critical receptor determinants for the hCoV-HKU1 infection. Interestingly, the virus seems to employ a type of sialic acid different from those employed by other group 2a CoVs. In addition, we determined that the HKU1-HE protein is an O-acetylesterase and acts as a receptor-destroying enzyme (RDE) for hCoV-HKU1. This is the first study to demonstrate that hCoV-HKU1 uses certain types of O-acetylated sialic acid residues on glycoproteins to initiate the infection of host cells and that the HKU1-HE protein possesses sialate-O-acetylesterase RDE activity.


Assuntos
Coronavirus/fisiologia , Hemaglutininas Virais/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Receptores Virais/análise , Glicoproteína da Espícula de Coronavírus/metabolismo , Proteínas Virais de Fusão/metabolismo , Ligação Viral , Células Cultivadas , Coronavirus/enzimologia , Humanos
11.
J Virol ; 89(11): 6093-104, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25833045

RESUMO

UNLABELLED: Coronaviruses (CoVs) and low-pathogenicity influenza A viruses (LP IAVs) depend on target cell proteases to cleave their viral glycoproteins and prime them for virus-cell membrane fusion. Several proteases cluster into tetraspanin-enriched microdomains (TEMs), suggesting that TEMs are preferred virus entry portals. Here we found that several CoV receptors and virus-priming proteases were indeed present in TEMs. Isolated TEMs, when mixed with CoV and LP IAV pseudoparticles, cleaved viral fusion proteins to fusion-primed fragments and potentiated viral transductions. That entering viruses utilize TEMs as a protease source was further confirmed using tetraspanin antibodies and tetraspanin short hairpin RNAs (shRNAs). Tetraspanin antibodies inhibited CoV and LP IAV infections, but their virus-blocking activities were overcome by expressing excess TEM-associated proteases. Similarly, cells with reduced levels of the tetraspanin CD9 resisted CoV pseudoparticle transductions but were made susceptible by overproducing TEM-associated proteases. These findings indicated that antibodies and CD9 depletions interfere with viral proteolytic priming in ways that are overcome by surplus proteases. TEMs appear to be exploited by some CoVs and LP IAVs for appropriate coengagement with cell receptors and proteases. IMPORTANCE: Enveloped viruses use their surface glycoproteins to catalyze membrane fusion, an essential cell entry step. Host cell components prime these viral surface glycoproteins to catalyze membrane fusion at specific times and places during virus cell entry. Among these priming components are proteases, which cleave viral surface glycoproteins, unleashing them to refold in ways that catalyze virus-cell membrane fusions. For some enveloped viruses, these proteases are known to reside on target cell surfaces. This research focuses on coronavirus and influenza A virus cell entry and identifies TEMs as sites of viral proteolysis, thereby defining subcellular locations of virus priming with greater precision. Implications of these findings extend to the use of virus entry antagonists, such as protease inhibitors, which might be most effective when localized to these microdomains.


Assuntos
Coronavirus/fisiologia , Vírus da Influenza A/fisiologia , Microdomínios da Membrana/virologia , Tetraspaninas/metabolismo , Ligação Viral , Internalização do Vírus , Animais , Humanos , Microdomínios da Membrana/química , Microdomínios da Membrana/metabolismo , Peptídeo Hidrolases/análise , Proteólise , Receptores Virais/análise , Proteínas Virais de Fusão/metabolismo
12.
J Virol ; 89(5): 2494-506, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25540384

RESUMO

UNLABELLED: Skeletal muscle, at 30 to 40% of body mass, is the most abundant soft tissue in the body. Besides its primary function in movement and posture, skeletal muscle is a significant innate immune organ with the capacity to produce cytokines and chemokines and respond to proinflammatory cytokines. Little is known about the role of skeletal muscle during systemic influenza A virus infection in any host and particularly avian species. Here we used primary chicken and duck multinucleated myotubes to examine their susceptibility and innate immune response to influenza virus infections. Both chicken and duck myotubes expressed avian and human sialic acid receptors and were readily susceptible to low-pathogenicity (H2N3 A/mallard duck/England/7277/06) and high-pathogenicity (H5N1 A/turkey/England/50-92/91 and H5N1 A/turkey/Turkey/1/05) avian and human H1N1 (A/USSR/77) influenza viruses. Both avian host species produced comparable levels of progeny H5N1 A/turkey/Turkey/1/05 virus. Notably, the rapid accumulation of viral nucleoprotein and matrix (M) gene RNA in chicken and duck myotubes was accompanied by extensive cytopathic damage with marked myotube apoptosis (widespread microscopic blebs, caspase 3/7 activation, and annexin V binding at the plasma membrane). Infected chicken myotubes produced significantly higher levels of proinflammatory cytokines than did the corresponding duck cells. Additionally, in chicken myotubes infected with H5N1 viruses, the induction of interferon beta (IFN-ß) and IFN-inducible genes, including the melanoma differentiation-associated protein 5 (MDA-5) gene, was relatively weak compared to infection with the corresponding H2N3 virus. Our findings highlight that avian skeletal muscle fibers are capable of productive influenza virus replication and are a potential tissue source of infection. IMPORTANCE: Infection with high-pathogenicity H5N1 viruses in ducks is often asymptomatic, and skeletal muscle from such birds could be a source of infection of humans and animals. Little is known about the ability of influenza A viruses to replicate in avian skeletal muscle fibers. We show here that cultured chicken and duck myotubes were highly susceptible to infection with both low- and high-pathogenicity avian influenza viruses. Infected myotubes of both avian species displayed rapid virus accumulation, apoptosis, and extensive cellular damage. Our results indicate that avian skeletal muscle fibers of chicken and duck could be significant contributors to progeny production of highly pathogenic H5N1 viruses.


Assuntos
Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H3N2/crescimento & desenvolvimento , Virus da Influenza A Subtipo H5N1/crescimento & desenvolvimento , Fibras Musculares Esqueléticas/virologia , Animais , Apoptose , Células Cultivadas , Galinhas , Citocinas/metabolismo , Efeito Citopatogênico Viral , Patos , Perfilação da Expressão Gênica , RNA Mensageiro/biossíntese , RNA Viral/biossíntese , Receptores Virais/análise , Ácidos Siálicos/análise
13.
Infect Immun ; 83(1): 28-38, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25312954

RESUMO

Shiga toxins (Stx) are a family of cytotoxic proteins that can cause hemolytic-uremic syndrome (HUS), a thrombotic microangiopathy, following infections by Shiga toxin-producing Escherichia coli (STEC). Renal failure is a key feature of HUS and a major cause of childhood renal failure worldwide. There are currently no specific therapies for STEC-associated HUS, and the mechanism of Stx-induced renal injury is not well understood primarily due to a lack of fully representative animal models and an inability to monitor disease progression on a molecular or cellular level in humans at early stages. Three-dimensional (3D) tissue models have been shown to be more in vivo-like in their phenotype and physiology than 2D cultures for numerous disease models, including cancer and polycystic kidney disease. It is unknown whether exposure of a 3D renal tissue model to Stx will yield a more in vivo-like response than 2D cell culture. In this study, we characterized Stx2-mediated cytotoxicity in a bioengineered 3D human renal tissue model previously shown to be a predictor of drug-induced nephrotoxicity and compared its response to Stx2 exposure in 2D cell culture. Our results demonstrate that although many mechanistic aspects of cytotoxicity were similar between 3D and 2D, treatment of the 3D tissues with Stx resulted in an elevated secretion of the kidney injury marker 1 (Kim-1) and the cytokine interleukin-8 compared to the 2D cell cultures. This study represents the first application of 3D tissues for the study of Stx-mediated kidney injury.


Assuntos
Rim/efeitos dos fármacos , Organoides/efeitos dos fármacos , Toxina Shiga II/toxicidade , Receptor Celular 1 do Vírus da Hepatite A , Humanos , Glicoproteínas de Membrana/análise , Modelos Biológicos , Técnicas de Cultura de Órgãos , Receptores Virais/análise
14.
J Virol ; 88(15): 8445-56, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24829344

RESUMO

UNLABELLED: Some animal influenza A viruses (IAVs) bind not only to N-acetylneuraminic acid (Neu5Ac) but also to N-glycolylneuraminic acid (Neu5Gc), which has been discussed as a virus receptor. Human cells cannot synthesize Neu5Gc due to dysfunction of the CMP-Neu5Ac hydroxylase (CMAH) gene, which converts CMP-Neu5Ac to CMP-Neu5Gc. However, exogenous Neu5Gc from Neu5Gc-rich dietary sources is able to be metabolically incorporated into surfaces of tissue cells and may be related to enhancement of the infectivity and severity of IAV. Here, we investigated the receptor function of Neu5Gc on IAV infection in Neu5Gc-expressing cells by transfection of the monkey CMAH gene into human cells or by incubation with human cells in the presence of N-glycolylmannosamine. Expression of Neu5Gc on human cells clearly suppressed infectivity of IAVs that possess Neu5Gc binding ability. Furthermore, there was no difference in infectivity of a transfectant virus that included the wild-type HA gene from A/Memphis/1/1971 (H3N2), which shows no Neu5Gc binding, between parent MCF7 cells and cells stably expressing the monkey CMAH gene (CMAH-MCF7 cells). On the other hand, cell entry of the transfectant virus that included the Neu5Gc-binding HA gene with a single mutation to Tyr at position Thr155 was arrested at the stage of internalization from the plasma membrane of the CMAH-MCF7 cells. These results indicate that expression of Neu5Gc on the surface of human epithelial cells suppresses infection of IAVs that possess Neu5Gc binding ability. Neu5Gc is suggested to work as a decoy receptor of Neu5Gc-binding IAVs but not a functional receptor for IAV infection. IMPORTANCE: Influenza A viruses (IAVs) bind to the host cell surfaces through sialic acids at the terminal of glycoconjugates. For IAV binding to sialic acids, some IAVs bind not only to N-acetylneuraminic acid (Neu5Ac) as a receptor but also to N-glycolylneuraminic acid (Neu5Gc). Neu5Gc has been discussed as a receptor of human and animal IAVs. Our results showed that Neu5Gc expression on human epithelial cells suppresses infection of IAVs that possess Neu5Gc binding ability. Neu5Gc is suggested to be a "decoy receptor" of Neu5Gc-binding IAVs but not a functional receptor for IAV infection. Human cells cannot synthesize Neu5Gc because of dysfunction of the CMP-N-acetylneuraminic acid hydroxylase gene but can exogenously and metabolically incorporate Neu5Gc from dietary sources. The expression of Neu5Gc on human epithelial cells by taking in exogenous Neu5Gc from Neu5Gc-rich dietary sources may be related to restriction of the infection of IAVs that have acquired Neu5Gc binding ability.


Assuntos
Membrana Celular/química , Células Epiteliais/química , Células Epiteliais/virologia , Vírus da Influenza A/fisiologia , Ácidos Neuramínicos/análise , Receptores Virais/análise , Internalização do Vírus , Animais , Linhagem Celular , Haplorrinos , Humanos
15.
Mol Cell Probes ; 29(2): 129-34, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25725345

RESUMO

The susceptibility of the host to influenza virus is determined by the distribution of the sialic acid (SA) receptors on host cell membrane. Avian influenza virus (AIV) preferentially binds to SA α-2,3-galactose (SA α2,3-gal) linked receptors, while human strains bind to sialic acid α2,6-galactose (SA α2,6-gal) linked receptors. Here, we describe the SA patterns and distributions in the reproductive tract of hens by employing two specific lectins, Maackia amurensis agglutinin (MAA) for SA α2,3-gal and sambucus nigra agglutinin (SNA) for SA α 2,6-gal receptors. Our results revealed that both SA α2,3-gal and SA α2,6-gal receptors exist in the reproductive tract of hens, including magnum, isthmus, uterus and vagina except for infundibulum. The distribution of SAα-2,3-gal receptor was more abundantly in the columnar epithelium cells of magnum, isthmus and uterus. Only minimal positive results for SA α-2,6-gal receptors were detected in the columnar epithelium cells of magnum, isthmus, uterus and vagina. Furthermore, AIV in tissues of the reproductive tract tissues of laying hens were detected by SYBR green-based reverse transcription and polymerase chain reaction (RT-PCR). Results showed that both viral loads and pathological changes in different parts of the reproductive tract were positively correlated with the expression of both receptors. Our results revealed that the reproductive tract of hens may provide an environment for the replication of both avian and human influenza viruses.


Assuntos
Galinhas/metabolismo , Influenza Aviária/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores Virais/análise , Animais , Células Epiteliais , Fito-Hemaglutininas/metabolismo , Reprodução , Sambucus nigra/metabolismo , Carga Viral
16.
J Infect Dis ; 210(2): 183-91, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24459192

RESUMO

BACKGROUND: Noroviruses (NoVs) represent a considerable public health burden. Despite their enormous genetic diversity, most outbreaks are due to the single GII.4 genotype, but the reasons for this are poorly understood. NoVs use histo-blood group antigens (HBGAs) as attachment factors. Since HBGAs are present in saliva, binding of strains to saliva is commonly used as a surrogate for recognition of the gut surface by specific strains, although the relationship between saliva and gut tissue expression of HBGAs is not well defined. METHODS: The presence of fucosylated HBGAs in saliva and stomach biopsy specimens, as well as that of genogroup I.1 and genogroup II.4 virus-like particles, were compared in a series of 109 donors from Portugal. RESULTS: An overall good concordance between HBGA expression in saliva and stomach surface mucosa was observed. However, unexpected mucosal expression of α(1,2)fucosylated epitopes in nonsecretor individuals was frequently detected, allowing for GII.4 attachment. Although all individuals were infected with Helicobacter pylori, abnormal expression of α(1,2)fucosylated motifs and binding of GII.4 virus-like particles in nonsecretors' mucosa were associated with positivity for the H. pylori CagA virulence factor. CONCLUSIONS: Infection by CagA-positive H. pylori induces expression of GII.4 attachment factors in nonsecretors' mucosa, expanding the host range of these strains and thereby possibly contributing to their epidemiological dominance.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Antígenos de Grupos Sanguíneos/análise , Mucosa Gástrica/química , Mucosa Gástrica/virologia , Norovirus/isolamento & purificação , Saliva/química , Saliva/virologia , Genótipo , Voluntários Saudáveis , Infecções por Helicobacter/microbiologia , Helicobacter pylori/isolamento & purificação , Especificidade de Hospedeiro , Interações Hospedeiro-Patógeno , Humanos , Norovirus/classificação , Norovirus/genética , Portugal , Receptores Virais/análise , Fatores de Virulência/metabolismo
17.
J Virol ; 87(4): 2036-45, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23192875

RESUMO

Animal influenza viruses (AIVs) are a major threat to human health and the source of pandemic influenza. A reliable small-mammal model to study the pathogenesis of infection and for testing vaccines and therapeutics against multiple strains of influenza virus is highly desirable. We show that cotton rats (Sigmodon hispidus) are susceptible to avian and swine influenza viruses. Cotton rats express α2,3-linked sialic acid (SA) and α2,6-linked SA residues in the trachea and α2,6-linked SA residues in the lung parenchyma. Prototypic avian influenza viruses (H3N2, H9N2, and H5N1) and swine-origin 2009 pandemic H1N1 viruses replicated in the nose and in the respiratory tract of cotton rats without prior adaptation and produced strong lung pathology that was characterized by early lung neutrophilia, followed by subsequent pneumonia. Consistent with other natural and animal models of influenza, only the H5N1 virus was lethal for cotton rats. More importantly, we show that the different avian and pandemic H1N1 strains tested are strong activators of the type I interferon (IFN)-inducible MX-1 gene both locally and systemically. Our data indicate that the cotton rat is a suitable small-mammal model to study the infection of animal influenza viruses and for validation of vaccines and therapeutics against these viruses.


Assuntos
Modelos Animais de Doenças , Vírus da Influenza A/patogenicidade , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Sigmodontinae/virologia , Animais , Vírus da Influenza A/crescimento & desenvolvimento , Pulmão/química , Pulmão/patologia , Pulmão/virologia , Receptores Virais/análise , Ácidos Siálicos/análise , Análise de Sobrevida , Traqueia/química , Traqueia/virologia
18.
Analyst ; 139(21): 5362-6, 2014 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-25192033

RESUMO

Urinary KIM-1 is an ideal biomarker for acute kidney injury diagnosis. The proof-of-concept is demonstrated by utilizing the hydroxyapatite derived from natural fish scales as an electrode material, where the sensing of KIM-1 is shown to be possible for the first time with a linear range from 0.01 to 0.20 µg mL(-1) and a detection limit of 0.017 µg mL(-1) under model conditions; proof-of-concept is demonstrated in spiked urine.


Assuntos
Biomarcadores/análise , Durapatita/química , Técnicas Eletroquímicas/instrumentação , Peixes , Glicoproteínas de Membrana/análise , Receptores Virais/análise , Animais , Receptor Celular 1 do Vírus da Hepatite A , Limite de Detecção
19.
Int J Immunopathol Pharmacol ; 27(4): 553-61, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25572735

RESUMO

Chemokines have been shown to play a critical role in tumor development and progression. However, little is known about the function and molecular mechanisms of CXCR6 in multiple malignancies. In the present study, we aimed to investigate the role of CXCR6 in human hepatocellular carcinoma (HCC). The expression of CXCR6 was examined by immunohistochemical assay using a tissue microarray procedure. A loss-of-function experiment was performed to explore the effects of lentivirus-mediated CXCR6 shRNA (shCXCR6) on cell proliferation and invasive potential by MTT and Transwell assays in HCC cell line (SMMC-7721). It was found that the expression of CXCR6 protein was significantly increased in HCC tissues compared with that in adjacent non-cancerous tissues (ANCT) (63.04% vs 36.96%, P=0.019), and correlated with the lymph-vascular space invasion in HCC patients (P=0.038). Knockdown of CXCR6 repressed cell proliferation and invasion of HCC cells followed by the down-regulation of vascular endothelial growth factor (VEGF). Taken together, our findings show that high expression of CXCR6 is positively associated with distant invasion of HCC patients, and blockade of CXCR6 signaling suppresses the growth and invasion of HCC cells through inhibition of the VEGF expression, suggesting that CXCR6 may represent a promising therapeutic target for the treatment of HCC.


Assuntos
Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Receptores de Quimiocinas/fisiologia , Receptores Virais/fisiologia , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Adulto , Idoso , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Receptores CXCR6 , Receptores de Quimiocinas/análise , Receptores de Quimiocinas/antagonistas & inibidores , Receptores Virais/análise , Receptores Virais/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA