Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
J Virol ; 98(4): e0010224, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38470058

RESUMEN

The transmembrane serine protease 2 (TMPRSS2) activates the outer structural proteins of a number of respiratory viruses including influenza A virus (IAV), parainfluenza viruses, and various coronaviruses for membrane fusion. Previous studies showed that TMPRSS2 interacts with the carboxypeptidase angiotensin-converting enzyme 2 (ACE2), a cell surface protein that serves as an entry receptor for some coronaviruses. Here, by using protease activity assays, we determine that ACE2 increases the enzymatic activity of TMPRSS2 in a non-catalytic manner. Furthermore, we demonstrate that ACE2 knockdown inhibits TMPRSS2-mediated cleavage of IAV hemagglutinin (HA) in Calu-3 human airway cells and suppresses virus titers 100- to 1.000-fold. Transient expression of ACE2 in ACE2-deficient cells increased TMPRSS2-mediated HA cleavage and IAV replication. ACE2 knockdown also reduced titers of MERS-CoV and prevented S cleavage by TMPRSS2 in Calu-3 cells. By contrast, proteolytic activation and multicycle replication of IAV with multibasic HA cleavage site typically cleaved by furin were not affected by ACE2 knockdown. Co-immunoprecipitation analysis revealed that ACE2-TMPRSS2 interaction requires the enzymatic activity of TMPRSS2 and the carboxypeptidase domain of ACE2. Together, our data identify ACE2 as a new co-factor or stabilizer of TMPRSS2 activity and as a novel host cell factor involved in proteolytic activation and spread of IAV in human airway cells. Furthermore, our data indicate that ACE2 is involved in the TMPRSS2-catalyzed activation of additional respiratory viruses including MERS-CoV.IMPORTANCEProteolytic cleavage of viral envelope proteins by host cell proteases is essential for the infectivity of many viruses and relevant proteases provide promising drug targets. The transmembrane serine protease 2 (TMPRSS2) has been identified as a major activating protease of several respiratory viruses, including influenza A virus. TMPRSS2 was previously shown to interact with angiotensin-converting enzyme 2 (ACE2). Here, we report the mechanistic details of this interaction. We demonstrate that ACE2 increases or stabilizes the enzymatic activity of TMPRSS2. Furthermore, we describe ACE2 involvement in TMPRSS2-catalyzed cleavage of the influenza A virus hemagglutinin and MERS-CoV spike protein in human airway cells. These findings expand our knowledge of the activation of respiratory viruses by TMPRSS2 and the host cell factors involved. In addition, our results could help to elucidate a physiological role for TMPRSS2.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Virus de la Influenza A , Pulmón , Proteolisis , Serina Endopeptidasas , Animales , Perros , Humanos , Enzima Convertidora de Angiotensina 2/deficiencia , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Biocatálisis , Línea Celular , Furina/metabolismo , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/metabolismo , Pulmón/citología , Pulmón/virología , Coronavirus del Síndrome Respiratorio de Oriente Medio/metabolismo , Unión Proteica , Serina Endopeptidasas/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus , Replicación Viral
2.
Sci Adv ; 8(40): eabm5859, 2022 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-36197970

RESUMEN

Molecular responses to influenza A virus (IAV) infections vary between mammalian species. To identify conserved and species-specific molecular responses, we perform a comparative study of transcriptomic data derived from blood cells, primary epithelial cells, and lung tissues collected from IAV-infected humans, ferrets, and mice. The molecular responses in the human host have unique functions such as antigen processing that are not observed in mice or ferrets. Highly conserved gene coexpression modules across the three species are enriched for IAV infection-induced pathways including cell cycle and interferon (IFN) signaling. TDRD7 is predicted as an IFN-inducible host factor that is up-regulated upon IAV infection in the three species. TDRD7 is required for antiviral IFN response, potentially modulating IFN signaling via the JAK/STAT/IRF9 pathway. Identification of the common and species-specific molecular signatures, networks, and regulators of IAV infection provides insights into host-defense mechanisms and will facilitate the development of novel therapeutic interventions against IAV infection.


Asunto(s)
Enfermedades Transmisibles , Virus de la Influenza A , Gripe Humana , Infecciones por Orthomyxoviridae , Animales , Antivirales , Hurones/metabolismo , Humanos , Virus de la Influenza A/fisiología , Gripe Humana/genética , Interferones/metabolismo , Ratones , Infecciones por Orthomyxoviridae/genética , Ribonucleoproteínas
3.
Nat Microbiol ; 6(10): 1319-1333, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34556855

RESUMEN

The fate of influenza A virus (IAV) infection in the host cell depends on the balance between cellular defence mechanisms and viral evasion strategies. To illuminate the landscape of IAV cellular restriction, we generated and integrated global genetic loss-of-function screens with transcriptomics and proteomics data. Our multi-omics analysis revealed a subset of both IFN-dependent and independent cellular defence mechanisms that inhibit IAV replication. Amongst these, the autophagy regulator TBC1 domain family member 5 (TBC1D5), which binds Rab7 to enable fusion of autophagosomes and lysosomes, was found to control IAV replication in vitro and in vivo and to promote lysosomal targeting of IAV M2 protein. Notably, IAV M2 was observed to abrogate TBC1D5-Rab7 binding through a physical interaction with TBC1D5 via its cytoplasmic tail. Our results provide evidence for the molecular mechanism utilised by IAV M2 protein to escape lysosomal degradation and traffic to the cell membrane, where it supports IAV budding and growth.


Asunto(s)
Autofagia , Evasión Inmune , Virus de la Influenza A/fisiología , Antivirales/metabolismo , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Interacciones Huésped-Patógeno , Humanos , Virus de la Influenza A/patogenicidad , Lisosomas/metabolismo , Unión Proteica , Proteínas de la Matriz Viral/metabolismo , Replicación Viral , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión a GTP rab7
4.
J Virol ; 95(20): e0090621, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34319155

RESUMEN

Cleavage of the influenza A virus (IAV) hemagglutinin (HA) by host proteases is indispensable for virus replication. Most IAVs possess a monobasic HA cleavage site cleaved by trypsin-like proteases. Previously, the transmembrane protease TMPRSS2 was shown to be essential for proteolytic activation of IAV HA subtypes H1, H2, H7, and H10 in mice. In contrast, additional proteases are involved in activation of certain H3 IAVs, indicating that HAs with monobasic cleavage sites can differ in their sensitivity to host proteases. Here, we investigated the role of TMPRSS2 in proteolytic activation of avian HA subtypes H1 to H11 and H14 to H16 in human and mouse airway cell cultures. Using reassortant viruses carrying representative HAs, we analyzed HA cleavage and multicycle replication in (i) lung cells of TMPRSS2-deficient mice and (ii) Calu-3 cells and primary human bronchial cells subjected to morpholino oligomer-mediated knockdown of TMPRSS2 activity. TMPRSS2 was found to be crucial for activation of H1 to H11, H14, and H15 in airway cells of human and mouse. Only H9 with an R-S-S-R cleavage site and H16 were proteolytically activated in the absence of TMPRSS2 activity, albeit with reduced efficiency. Moreover, a TMPRSS2-orthologous protease from duck supported activation of H1 to H11, H15, and H16 in MDCK cells. Together, our data demonstrate that in human and murine respiratory cells, TMPRSS2 is the major activating protease of almost all IAV HA subtypes with monobasic cleavage sites. Furthermore, our results suggest that TMPRSS2 supports activation of IAV with a monobasic cleavage site in ducks. IMPORTANCE Human infections with avian influenza A viruses upon exposure to infected birds are frequently reported and have received attention as a potential pandemic threat. Cleavage of the envelope glycoprotein hemagglutinin (HA) by host proteases is a prerequisite for membrane fusion and essential for virus infectivity. In this study, we identify the transmembrane protease TMPRSS2 as the major activating protease of avian influenza virus HAs of subtypes H1 to H11, H14 and H15 in human and murine airway cells. Our data demonstrate that inhibition of TMPRSS2 activity may provide a useful approach for the treatment of human infections with avian influenza viruses that should be considered for pandemic preparedness as well. Additionally, we show that a TMPRSS2-orthologous protease from duck can activate avian influenza virus HAs with a monobasic cleavage site and, thus, represents a potential virus-activating protease in waterfowl, the primary reservoir for influenza A viruses.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Virus de la Influenza A/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Bronquios/citología , Línea Celular , Perros , Femenino , Células HEK293 , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Hemaglutininas Virales/genética , Hemaglutininas Virales/metabolismo , Interacciones Huésped-Patógeno , Humanos , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H3N2 del Virus de la Influenza A/fisiología , Virus de la Influenza A/inmunología , Virus de la Influenza A/patogenicidad , Pulmón/virología , Células de Riñón Canino Madin Darby , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Péptido Hidrolasas/metabolismo , Proteolisis , Mucosa Respiratoria/metabolismo , Serina Endopeptidasas/fisiología , Replicación Viral
5.
J Antimicrob Chemother ; 76(2): 413-417, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33164048

RESUMEN

BACKGROUND: As the causative agent of COVID-19, SARS-CoV-2 is a pathogen of immense importance to global public health. Development of innovative direct-acting antiviral agents is sorely needed to address this virus. Peptide-conjugated morpholino oligomers (PPMO) are antisense compounds composed of a phosphorodiamidate morpholino oligomer covalently conjugated to a cell-penetrating peptide. PPMO require no delivery assistance to enter cells and are able to reduce expression of targeted RNA through sequence-specific steric blocking. METHODS: Five PPMO designed against sequences of genomic RNA in the SARS-CoV-2 5'-untranslated region and a negative control PPMO of random sequence were synthesized. Each PPMO was evaluated for its effect on the viability of uninfected cells and its inhibitory effect on the replication of SARS-CoV-2 in Vero-E6 cell cultures. Cell viability was evaluated with an ATP-based method using a 48 h PPMO treatment time. Viral growth was measured with quantitative RT-PCR and TCID50 infectivity assays from experiments where cells received a 5 h PPMO treatment time. RESULTS: PPMO designed to base-pair with sequence in the 5' terminal region or the leader transcription regulatory sequence region of SARS-CoV-2 genomic RNA were highly efficacious, reducing viral titres by up to 4-6 log10 in cell cultures at 48-72 h post-infection, in a non-toxic and dose-responsive manner. CONCLUSIONS: The data indicate that PPMO have the ability to potently and specifically suppress SARS-CoV-2 growth and are promising candidates for further preclinical development.


Asunto(s)
Antivirales/farmacología , COVID-19/virología , Péptidos de Penetración Celular/farmacología , Morfolinos/farmacología , SARS-CoV-2/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Antivirales/química , Supervivencia Celular/efectos de los fármacos , Péptidos de Penetración Celular/química , Chlorocebus aethiops , Efecto Citopatogénico Viral/efectos de los fármacos , Morfolinos/química , SARS-CoV-2/genética , Células Vero
6.
bioRxiv ; 2020 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-33024974

RESUMEN

BACKGROUND: SARS-CoV-2 is the causative agent of COVID-19 and a pathogen of immense global public health importance. Development of innovative direct-acting antiviral agents is sorely needed to address this virus. Peptide-conjugated morpholino oligomers (PPMO) are antisense agents composed of a phosphordiamidate morpholino oligomer covalently conjugated to a cell-penetrating peptide. PPMO require no delivery assistance to enter cells and are able to reduce expression of targeted RNA through sequence-specific steric blocking. OBJECTIVES AND METHODS: Five PPMO designed against sequences of genomic RNA in the SARS-CoV-2 5'-untranslated region and a negative control PPMO of random sequence were synthesized. Each PPMO was evaluated for its effect on the viability of uninfected cells and its inhibitory effect on the replication of SARS-CoV-2 in Vero-E6 cell cultures. Cell viability was evaluated with an ATP-based method and viral growth was measured with quantitative RT-PCR and TCID 50 infectivity assays. RESULTS: PPMO designed to base-pair with sequence in the 5'-terminal region or the leader transcription regulatory sequence-region of SARS-CoV-2 genomic RNA were highly efficacious, reducing viral titers by up to 4-6 log10 in cell cultures at 48-72 hours post-infection, in a non-toxic and dose-responsive manner. CONCLUSION: The data indicate that PPMO have the ability to potently and specifically suppress SARS-CoV-2 growth and are promising candidates for further pre-clinical development.

7.
Life Sci Alliance ; 3(9)2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32703818

RESUMEN

The novel emerged SARS-CoV-2 has rapidly spread around the world causing acute infection of the respiratory tract (COVID-19) that can result in severe disease and lethality. For SARS-CoV-2 to enter cells, its surface glycoprotein spike (S) must be cleaved at two different sites by host cell proteases, which therefore represent potential drug targets. In the present study, we show that S can be cleaved by the proprotein convertase furin at the S1/S2 site and the transmembrane serine protease 2 (TMPRSS2) at the S2' site. We demonstrate that TMPRSS2 is essential for activation of SARS-CoV-2 S in Calu-3 human airway epithelial cells through antisense-mediated knockdown of TMPRSS2 expression. Furthermore, SARS-CoV-2 replication was also strongly inhibited by the synthetic furin inhibitor MI-1851 in human airway cells. In contrast, inhibition of endosomal cathepsins by E64d did not affect virus replication. Combining various TMPRSS2 inhibitors with furin inhibitor MI-1851 produced more potent antiviral activity against SARS-CoV-2 than an equimolar amount of any single serine protease inhibitor. Therefore, this approach has considerable therapeutic potential for treatment of COVID-19.


Asunto(s)
Células Epiteliales Alveolares/virología , Betacoronavirus/fisiología , Furina/genética , Serina Endopeptidasas/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Células Epiteliales Alveolares/citología , Animales , Sitios de Unión , Línea Celular , Chlorocebus aethiops , Células HEK293 , Humanos , Proteolisis , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/química , Células Vero , Internalización del Virus , Replicación Viral
8.
J Virol ; 93(21)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31391268

RESUMEN

Cleavage of influenza virus hemagglutinin (HA) by host cell proteases is essential for virus infectivity and spread. We previously demonstrated in vitro that the transmembrane protease TMPRSS2 cleaves influenza A virus (IAV) and influenza B virus (IBV) HA possessing a monobasic cleavage site. Subsequent studies revealed that TMPRSS2 is crucial for the activation and pathogenesis of H1N1pdm and H7N9 IAV in mice. In contrast, activation of H3N2 IAV and IBV was found to be independent of TMPRSS2 expression and supported by an as-yet-undetermined protease(s). Here, we investigated the role of TMPRSS2 in proteolytic activation of IAV and IBV in three human airway cell culture systems: primary human bronchial epithelial cells (HBEC), primary type II alveolar epithelial cells (AECII), and Calu-3 cells. Knockdown of TMPRSS2 expression was performed using a previously described antisense peptide-conjugated phosphorodiamidate morpholino oligomer, T-ex5, that interferes with splicing of TMPRSS2 pre-mRNA, resulting in the expression of enzymatically inactive TMPRSS2. T-ex5 treatment produced efficient knockdown of active TMPRSS2 in all three airway cell culture models and prevented proteolytic activation and multiplication of H7N9 IAV in Calu-3 cells and H1N1pdm, H7N9, and H3N2 IAV in HBEC and AECII. T-ex5 treatment also inhibited the activation and spread of IBV in AECII but did not affect IBV activation in HBEC and Calu-3 cells. This study identifies TMPRSS2 as the major HA-activating protease of IAV in human airway cells and IBV in type II pneumocytes and as a potential target for the development of novel drugs to treat influenza infections.IMPORTANCE Influenza A viruses (IAV) and influenza B viruses (IBV) cause significant morbidity and mortality during seasonal outbreaks. Cleavage of the viral surface glycoprotein hemagglutinin (HA) by host proteases is a prerequisite for membrane fusion and essential for virus infectivity. Inhibition of relevant proteases provides a promising therapeutic approach that may avoid the development of drug resistance. HA of most influenza viruses is cleaved at a monobasic cleavage site, and a number of proteases have been shown to cleave HA in vitro This study demonstrates that the transmembrane protease TMPRSS2 is the major HA-activating protease of IAV in primary human bronchial cells and of both IAV and IBV in primary human type II pneumocytes. It further reveals that human and murine airway cells can differ in their HA-cleaving protease repertoires. Our data will help drive the development of potent and selective protease inhibitors as novel drugs for influenza treatment.


Asunto(s)
Virus de la Influenza A/fisiología , Virus de la Influenza B/fisiología , Gripe Humana/virología , Serina Endopeptidasas/metabolismo , Animales , Bronquios/citología , Células Cultivadas , Células Epiteliales/virología , Técnicas de Silenciamiento del Gen , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Interacciones Huésped-Patógeno , Humanos , Gripe Humana/enzimología , Gripe Humana/metabolismo , Ratones , Infecciones por Orthomyxoviridae/enzimología , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Alveolos Pulmonares/citología , Serina Endopeptidasas/genética , Regulación hacia Arriba , Replicación Viral
9.
mBio ; 9(5)2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30181255

RESUMEN

RNA viruses that replicate in the cell cytoplasm typically concentrate their replication machinery within specialized compartments. This concentration favors enzymatic reactions and shields viral RNA from detection by cytosolic pattern recognition receptors. Nonsegmented negative-strand (NNS) RNA viruses, which include some of the most significant human, animal, and plant pathogens extant, form inclusions that are sites of RNA synthesis and are not circumscribed by a membrane. These inclusions share similarities with cellular protein/RNA structures such as P granules and nucleoli, which are phase-separated liquid compartments. Here we show that replication compartments of vesicular stomatitis virus (VSV) have the properties of liquid-like compartments that form by phase separation. Expression of the individual viral components of the replication machinery in cells demonstrates that the 3 viral proteins required for replication are sufficient to drive cytoplasmic phase separation. Therefore, liquid-liquid phase separation, previously linked to organization of P granules, nucleolus homeostasis, and cell signaling, plays a key role in host-pathogen interactions. This work suggests novel therapeutic approaches to the problem of combating NNS RNA viral infections.IMPORTANCE RNA viruses compartmentalize their replication machinery to evade detection by host pattern recognition receptors and concentrate the machinery of RNA synthesis. For positive-strand RNA viruses, RNA replication occurs in a virus-induced membrane-associated replication organelle. For NNS RNA viruses, the replication compartment is a cytoplasmic inclusion that is not circumscribed by a cellular membrane. Such structures were first observed in the cell bodies of neurons from humans infected with rabies virus and were termed Negri bodies. How the replication machinery that forms this inclusion remains associated in the absence of a membrane has been an enduring mystery. In this article, we present evidence that the VSV replication compartments form through phase separation. Phase separation is increasingly recognized as responsible for cellular structures as diverse as processing bodies (P-bodies) and nucleoli and was recently demonstrated for rabies virus. This article further links the fields of host-pathogen interaction with that of phase separation.


Asunto(s)
Gránulos Citoplasmáticos/virología , Interacciones Microbiota-Huesped , Cuerpos de Inclusión Viral/fisiología , Vesiculovirus/fisiología , Proteínas Virales/metabolismo , Replicación Viral , Animales , Compartimento Celular , Línea Celular , Chlorocebus aethiops , ARN Viral , Células Vero , Proteínas Virales/genética
10.
BMC Res Notes ; 11(1): 423, 2018 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-29970132

RESUMEN

OBJECTIVE: The objective of this study was to obtain clinical, virological and demographic data detailing the 2016 dengue outbreak in Nepal. RESULTS: Dengue disease was first reported in Nepal in 2004 and several major outbreaks have occurred since then, with a significant impact on public health. An outbreak of dengue fever occurred in Nepal during June to November 2016, with a peak number of cases reported in September. 1473 patients with laboratory confirmed DENV infections visited or were admitted to hospitals during this period. The most common clinical symptoms included fever, headache, joint pain and thrombocytopenia. Serotyping of 75 serum samples from patients having fever for less than 4 days was carried out with a dengue virus (DENV) serotype-specific RT-PCR strategy. Our results indicate that the dengue outbreak in Nepal during 2016 was caused predominantly, if not exclusively, by DENV-1, representing a shift in the prevailing serotype from DENV-2, the dominant serotype characterizing the 2013 dengue epidemic in Nepal. Hopefully, this report will assist Nepalese public health agencies in developing improved dengue-related programs including mosquito-vector control, DENV surveillance, and diagnosis and treatment of dengue fever patients, in order to reduce the impact of future dengue epidemics.


Asunto(s)
Dengue/epidemiología , Brotes de Enfermedades , Adulto , Animales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Nepal/epidemiología , Adulto Joven
11.
Biomed Res Int ; 2018: 9241679, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29487874

RESUMEN

HBV and HCV infections are widespread among the HIV-infected individuals in Nepal. The goals of this study were to investigate the epidemiological profile and risk factors for acquiring HBV and/or HCV coinfection in disadvantaged HIV-positive population groups in Nepal. We conducted a retrospective study on blood samples from HIV-positive patients from the National Public Health Laboratory at Kathmandu to assay for HBsAg, HBeAg, and anti-HCV antibodies, HIV viral load, and CD4+ T cell count. Among 579 subjects, the prevalence of HIV-HBV, HIV-HCV, and HIV-HBV-HCV coinfections was 3.62%, 2.93%, and 0.34%, respectively. Multivariate regression analysis indicated that spouses of HIV-positive migrant labourers were at significant risk for coinfection with HBV infection, and an age of >40 years in HIV-infected individuals was identified as a significant risk factor for HCV coinfection. Overall our study indicates that disadvantaged population groups such as intravenous drug users, migrant workers and their spouses, female sex workers, and men who have sex with HIV-infected men are at a high and persistent risk of acquiring viral hepatitis. We conclude that Nepalese HIV patients should receive HBV and HCV diagnostic screening on a regular basis.


Asunto(s)
Coinfección/epidemiología , Infecciones por VIH/complicaciones , Infecciones por VIH/epidemiología , Hepatitis B/epidemiología , Hepatitis B/etiología , Hepatitis C/epidemiología , Hepatitis C/etiología , Adulto , Linfocitos T CD4-Positivos/inmunología , Coinfección/sangre , Coinfección/inmunología , Femenino , Infecciones por VIH/sangre , Infecciones por VIH/inmunología , Hepacivirus/inmunología , Hepatitis B/sangre , Hepatitis B/inmunología , Antígenos de Superficie de la Hepatitis B/sangre , Antígenos e de la Hepatitis B/sangre , Virus de la Hepatitis B/inmunología , Hepatitis C/inmunología , Anticuerpos contra la Hepatitis C/sangre , Humanos , Masculino , Persona de Mediana Edad , Nepal/epidemiología , Estudios Retrospectivos , Factores de Riesgo , Adulto Joven
12.
Infect Agent Cancer ; 13: 4, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29375654

RESUMEN

BACKGROUND: Cervical cancer (CC) is the leading cause of morbidity and mortality from cancer in Nepalese women. Nearly all cases of CC are caused by infection with certain genotypes of human papillomavirus (HPV). Data on HPV genotype distribution in Nepalese CC patients is sparse. We aimed to determine the distribution of HPV genotypes in biopsies of CC tissue from Nepalese women. METHODS: This study examined 248 archived paraffin-embedded tissue specimens from CC cases from patients of B.P. Koirala Memorial Cancer Hospital, Bharatpur, Chitwan, Nepal. DNA was extracted from the biopsies and HPV detection performed by PCR. HPV genotyping was then carried out by a reverse line hybridization technique capable of identifying 36 distinct HPV genotypes. RESULTS: Most of the samples were from tumors that had been designated by hospital pathologists as squamous cell carcinoma (77.6%). 165 of the 248 samples contained DNA of sufficient quality for rigorous PCR testing. All the analyzable specimens were positive for HPV. The most common HPV genotypes, in decreasing order of frequency were 16, 18, 45, 33, 52, 56 and 31; most were found as single infections (94.5%). Together, HPV types 16, 18, and 45 were found in 92% of the tumor samples. CONCLUSION: This study strengthens the knowledge-base of HPV genotype distribution in CC cases in Nepal. Hopefully, this information will be useful to the medical community and public health policy-makers in generating improved HPV-surveillance, -prevention and -treatment strategies in Nepal.

13.
Nat Microbiol ; 2: 17022, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-28248290

RESUMEN

Retinoic acid-inducible gene I (RIG-I) receptor recognizes 5'-triphosphorylated RNA and triggers a signalling cascade that results in the induction of type-I interferon (IFN)-dependent responses. Its precise regulation represents a pivotal balance between antiviral defences and autoimmunity. To elucidate the cellular cofactors that regulate RIG-I signalling, we performed two global RNA interference analyses to identify both positive and negative regulatory nodes operating on the signalling pathway during virus infection. These factors were integrated with experimentally and computationally derived interactome data to build a RIG-I protein interaction network. Our analysis revealed diverse cellular processes, including the unfolded protein response, Wnt signalling and RNA metabolism, as critical cellular components governing innate responses to non-self RNA species. Importantly, we identified K-Homology Splicing Regulatory Protein (KHSRP) as a negative regulator of this pathway. We find that KHSRP associates with the regulatory domain of RIG-I to maintain the receptor in an inactive state and attenuate its sensing of viral RNA (vRNA). Consistent with increased RIG-I antiviral signalling in the absence of KHSRP, viral replication is reduced when KHSRP expression is knocked down both in vitro and in vivo. Taken together, these data indicate that KHSRP functions as a checkpoint regulator of the innate immune response to pathogen challenge.


Asunto(s)
Proteína 58 DEAD Box/antagonistas & inhibidores , ARN Viral/inmunología , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Células HEK293 , Humanos , Inmunidad Innata , Subtipo H1N1 del Virus de la Influenza A/inmunología , Unión Proteica , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Receptores Inmunológicos
14.
Mov Disord ; 31(4): 538-46, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26695437

RESUMEN

BACKGROUND: Continuous administration of levodopa-carbidopa intestinal gel (carbidopa-levodopa enteral suspension) through a percutaneous endoscopic gastrojejunostomy is a treatment option for advanced Parkinson disease (PD) patients with motor fluctuations resistant to standard oral medications. Safety data from 4 prospective studies were integrated to assess the safety of this therapy. METHODS: Safety data from 4 studies were summarized using 2 overlapping data sets, permitting the separation of procedure/device-associated (n = 395) from non-procedure/device adverse events (n = 412). RESULTS: At the data cutoff, median exposure to levodopa-carbidopa intestinal gel was 911 days (range, 1-1980 days) with 963 total patient-years of exposure. Procedure/device adverse events occurred in 300 patients (76%), and serious adverse events occurred in 68 (17%); most frequently reported procedure/device adverse events and serious adverse events were complications of device insertion (41% and 8%, respectively) and abdominal pain (36% and 4%, respectively). Non-procedure/device adverse events occurred in 92% (379), with most frequently reported being insomnia (23%) and falls (23%); 42% (171) had non-procedure/device serious adverse events, with most frequently reported being pneumonia (5%) and PD symptoms (2%). Adverse events led to discontinuation in 17% (72), most frequently because of complication of device insertion (2.4%). There were 34 treatment-emergent deaths (8.3%) in the overlapping data sets, 2 of which (0.5%) were considered "possibly related" to the treatment system. CONCLUSION: In the largest collection of levodopa-carbidopa intestinal gel safety data from prospective clinical studies, procedure/device events were frequently reported and occasionally life threatening. Most non-procedure/device events were typical for levodopa treatment and an elderly population. These factors combined with high treatment efficacy led to a relatively low discontinuation rate in advanced PD patients.


Asunto(s)
Antiparkinsonianos/efectos adversos , Carbidopa/efectos adversos , Ensayos Clínicos Fase III como Asunto/estadística & datos numéricos , Derivación Gástrica/efectos adversos , Infusiones Parenterales/efectos adversos , Levodopa/efectos adversos , Evaluación de Resultado en la Atención de Salud/estadística & datos numéricos , Enfermedad de Parkinson/tratamiento farmacológico , Anciano , Antiparkinsonianos/administración & dosificación , Carbidopa/administración & dosificación , Combinación de Medicamentos , Femenino , Geles , Humanos , Levodopa/administración & dosificación , Masculino , Persona de Mediana Edad , Estudios Multicéntricos como Asunto/estadística & datos numéricos , Estudios Prospectivos
15.
Cell Host Microbe ; 18(6): 723-35, 2015 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-26651948

RESUMEN

Several systems-level datasets designed to dissect host-pathogen interactions during influenza A infection have been reported. However, apparent discordance among these data has hampered their full utility toward advancing mechanistic and therapeutic knowledge. To collectively reconcile these datasets, we performed a meta-analysis of data from eight published RNAi screens and integrated these data with three protein interaction datasets, including one generated within the context of this study. Further integration of these data with global virus-host interaction analyses revealed a functionally validated biochemical landscape of the influenza-host interface, which can be queried through a simplified and customizable web portal (http://www.metascape.org/IAV). Follow-up studies revealed that the putative ubiquitin ligase UBR4 associates with the viral M2 protein and promotes apical transport of viral proteins. Taken together, the integrative analysis of influenza OMICs datasets illuminates a viral-host network of high-confidence human proteins that are essential for influenza A virus replication.


Asunto(s)
Proteínas de Unión a Calmodulina/metabolismo , Proteínas del Citoesqueleto/metabolismo , Interacciones Huésped-Patógeno , Virus de la Influenza A/fisiología , Proteínas de la Matriz Viral/metabolismo , Liberación del Virus , Animales , Línea Celular , Biología Computacional , Citometría de Flujo , Humanos , Inmunoprecipitación , Ratones Endogámicos BALB C , Microscopía Fluorescente , Unión Proteica , Mapas de Interacción de Proteínas , Transporte de Proteínas , Ubiquitina-Proteína Ligasas
16.
Antiviral Res ; 120: 134-9, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26086884

RESUMEN

Hepatitis E virus (HEV) infection is a cause of hepatitis in humans worldwide and has been associated with a case-fatality rate of up to 30% in pregnant women. Recently, persistent and chronic HEV infections have been recognized as a serious clinical problem, especially in immunocompromised individuals. To date, there are no FDA-approved HEV-specific antiviral drugs. In this study, we evaluated antisense peptide-conjugated morpholino oligomers (PPMO) designed against HEV genomic sequences as potential HEV-specific antiviral compounds. Two genetically-distinct strains of human HEV, genotype 1 Sar55 and genotype 3 Kernow-C1, isolated from patients with acute and chronic hepatitis, respectively, were used to evaluate inhibition of viral replication by PPMO in liver cells. The anti-HEV PPMO produced a significant reduction in the levels of HEV RNA and capsid protein, indicating effective inhibition of HEV replication. PPMO HP1, which targets a highly conserved sequence in the start site region of ORF1, was also effective against the genotype 3 Kernow-C1 strain in stably-infected HepG2/C3A liver cells. The antiviral activity observed was specific, dose-responsive and potent, suggesting that further exploration of PPMO HP1 as a potential HEV-specific antiviral agent is warranted.


Asunto(s)
Antivirales/farmacología , Portadores de Fármacos/metabolismo , Virus de la Hepatitis E/efectos de los fármacos , Morfolinos/farmacología , Oligonucleótidos Antisentido/farmacología , Péptidos/metabolismo , Replicación Viral/efectos de los fármacos , Proteínas de la Cápside/análisis , Línea Celular , Virus de la Hepatitis E/fisiología , Hepatocitos/virología , Humanos , Pruebas de Sensibilidad Microbiana , ARN Viral/análisis , Carga Viral
17.
Immunity ; 40(6): 880-95, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-24882218

RESUMEN

Type I interferons (IFN-I) are essential antiviral cytokines produced upon microbial infection. IFN-I elicits this activity through the upregulation of hundreds of IFN-I-stimulated genes (ISGs). The full breadth of ISG induction demands activation of a number of cellular factors including the IκB kinase epsilon (IKKε). However, the mechanism of IKKε activation upon IFN receptor signaling has remained elusive. Here we show that TRIM6, a member of the E3-ubiquitin ligase tripartite motif (TRIM) family of proteins, interacted with IKKε and promoted induction of IKKε-dependent ISGs. TRIM6 and the E2-ubiquitin conjugase UbE2K cooperated in the synthesis of unanchored K48-linked polyubiquitin chains, which activated IKKε for subsequent STAT1 phosphorylation. Our work attributes a previously unrecognized activating role of K48-linked unanchored polyubiquitin chains in kinase activation and identifies the UbE2K-TRIM6-ubiquitin axis as critical for IFN signaling and antiviral response.


Asunto(s)
Quinasa I-kappa B/inmunología , Interferón Tipo I/inmunología , Poliubiquitina/biosíntesis , Ubiquitina-Proteína Ligasas/inmunología , Animales , Antivirales , Células Cultivadas , Activación Enzimática/inmunología , Humanos , Janus Quinasa 1 , Ratones , Fosforilación/inmunología , Interferencia de ARN , ARN Interferente Pequeño , Factor de Transcripción STAT1/inmunología , Transducción de Señal/inmunología , Proteínas de Motivos Tripartitos , Enzimas Ubiquitina-Conjugadoras/inmunología , Ubiquitina-Proteína Ligasas/genética
18.
J Virol ; 88(10): 5533-42, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24599995

RESUMEN

UNLABELLED: Dengue viruses (DENV) are endemic pathogens of tropical and subtropical regions that cause significant morbidity and mortality worldwide. To date, no vaccines or antiviral therapeutics have been approved for combating DENV-associated disease. In this paper, we describe a class of tricyclic small-molecule compounds-dihydrodibenzothiepines (DHBTs), identified through high-throughput screening-with potent inhibitory activity against DENV serotype 2. SKI-417616, a highly active representative of this class, displayed activity against all four serotypes of DENV, as well as against a related flavivirus, West Nile virus (WNV), and an alphavirus, Sindbis virus (SINV). This compound was characterized to determine its mechanism of antiviral activity. Investigation of the stage of the viral life cycle affected revealed that an early event in the life cycle is inhibited. Due to the structural similarity of the DHBTs to known antagonists of the dopamine and serotonin receptors, we explored the roles of two of these receptors, serotonin receptor 2A (5HTR2A) and the D4 dopamine receptor (DRD4), in DENV infection. Antagonism of DRD4 and subsequent downstream phosphorylation of epidermal growth factor receptor (EGFR)-related kinase (ERK) were found to impact DENV infection negatively, and blockade of signaling through this network was confirmed as the mechanism of anti-DENV activity for this class of compounds. IMPORTANCE: The dengue viruses are mosquito-borne, reemerging human pathogens that are the etiological agents of a spectrum of febrile diseases. Currently, there are no approved therapeutic treatments for dengue-associated disease, nor is there a vaccine. This study identifies a small molecule, SKI-417616, with potent anti-dengue virus activity. Further analysis revealed that SKI-417616 acts through antagonism of the host cell dopamine D4 receptor and subsequent repression of the ERK phosphorylation pathway. These results suggest that SKI-417616, or other compounds targeting the same cellular pathways, may have therapeutic potential for the treatment of dengue virus infections.


Asunto(s)
Antivirales/metabolismo , Virus del Dengue/efectos de los fármacos , Virus del Dengue/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Receptores de Dopamina D4/antagonistas & inhibidores , Transducción de Señal , Replicación Viral/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Ensayos Analíticos de Alto Rendimiento , Humanos , Virus Sindbis/efectos de los fármacos , Virus Sindbis/fisiología , Virus del Nilo Occidental/efectos de los fármacos , Virus del Nilo Occidental/fisiología
19.
J Virol ; 87(17): 9411-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23824813

RESUMEN

Dengue virus has emerged as a global health threat to over one-third of humankind. As a positive-strand RNA virus, dengue virus relies on the host cell metabolism for its translation, replication, and egress. Therefore, a better understanding of the host cell metabolic pathways required for dengue virus infection offers the opportunity to develop new approaches for therapeutic intervention. In a recently described screen of known drugs and bioactive molecules, we observed that methotrexate and floxuridine inhibited dengue virus infections at low micromolar concentrations. Here, we demonstrate that all serotypes of dengue virus, as well as West Nile virus, are highly sensitive to both methotrexate and floxuridine, whereas other RNA viruses (Sindbis virus and vesicular stomatitis virus) are not. Interestingly, flavivirus replication was restored by folinic acid, a thymidine precursor, in the presence of methotrexate and by thymidine in the presence of floxuridine, suggesting an unexpected role for thymidine in flavivirus replication. Since thymidine is not incorporated into RNA genomes, it is likely that increased thymidine production is indirectly involved in flavivirus replication. A possible mechanism is suggested by the finding that p53 inhibition restored dengue virus replication in the presence of floxuridine, consistent with thymidine-less stress triggering p53-mediated antiflavivirus effects in infected cells. Our data reveal thymidine synthesis pathways as new and unexpected therapeutic targets for antiflaviviral drug development.


Asunto(s)
Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Virus del Dengue/metabolismo , Flavivirus/efectos de los fármacos , Flavivirus/metabolismo , Timidina/biosíntesis , Animales , Línea Celular , Chlorocebus aethiops , Virus ADN/efectos de los fármacos , Virus del Dengue/fisiología , Modelos Animales de Enfermedad , Flavivirus/fisiología , Infecciones por Flavivirus/tratamiento farmacológico , Floxuridina/farmacología , Células HEK293 , Células HeLa , Humanos , Leucovorina/farmacología , Metotrexato/farmacología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Virus ARN/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Células Vero , Replicación Viral/efectos de los fármacos , Virus del Nilo Occidental/efectos de los fármacos , Virus del Nilo Occidental/metabolismo , Virus del Nilo Occidental/fisiología
20.
Antimicrob Agents Chemother ; 55(10): 4631-8, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21825302

RESUMEN

Members of the Arenaviridae family are a threat to public health and can cause meningitis and hemorrhagic fever, and yet treatment options remain limited by a lack of effective antivirals. In this study, we found that peptide-conjugated phosphorodiamidate morpholino oligomers (PPMO) complementary to viral genomic RNA were effective in reducing arenavirus replication in cell cultures and in vivo. PPMO complementary to the Junín virus genome were designed to interfere with viral RNA synthesis or translation or both. However, only PPMO designed to potentially interfere with translation were effective in reducing virus replication. PPMO complementary to sequences that are highly conserved across the arenaviruses and located at the 5' termini of both genomic segments were effective against Junín virus, Tacaribe virus, Pichinde virus, and lymphocytic choriomeningitis virus (LCMV)-infected cell cultures and suppressed viral titers in the livers of LCMV-infected mice. These results suggest that arenavirus 5' genomic termini represent promising targets for pan-arenavirus antiviral therapeutic development.


Asunto(s)
Antivirales/farmacología , Arenavirus/efectos de los fármacos , Morfolinos/farmacología , Péptidos/farmacología , Animales , Infecciones por Arenaviridae/tratamiento farmacológico , Infecciones por Arenaviridae/virología , Arenavirus/genética , Arenavirus/crecimiento & desarrollo , Arenavirus del Nuevo Mundo/efectos de los fármacos , Línea Celular , Chlorocebus aethiops , Genoma Viral , Virus Junin/efectos de los fármacos , Virus de la Coriomeningitis Linfocítica/efectos de los fármacos , Ratones , Pruebas de Sensibilidad Microbiana , Virus Pichinde/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , ARN Viral/genética , Células Vero , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...