Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Oncoimmunology ; 13(1): 2377830, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39005546

RESUMO

Attenuated measles virus (MV) exerts its oncolytic activity in malignant pleural mesothelioma (MPM) cells that lack type-I interferon (IFN-I) production or responsiveness. However, other cells in the tumor microenvironment (TME), such as myeloid cells, possess functional antiviral pathways. In this study, we aimed to characterize the interplay between MV and the myeloid cells in human MPM. We cocultured MPM cell lines with monocytes or macrophages and infected them with MV. We analyzed the transcriptome of each cell type and studied their secretion and phenotypes by high-dimensional flow cytometry. We also measured transgene expression using an MV encoding GFP (MV-GFP). We show that MPM cells drive the differentiation of monocytes into M2-like macrophages. These macrophages inhibit GFP expression in tumor cells harboring a defect in IFN-I production and a functional signaling downstream of the IFN-I receptor, while having minimal effects on GFP expression in tumor cells with defect of responsiveness to IFN-I. Interestingly, inhibition of the IFN-I signaling by ruxolitinib restores GFP expression in tumor cells. Upon MV infection, cocultured macrophages express antiviral pro-inflammatory genes and induce the expression of IFN-stimulated genes in tumor cells. MV also increases the expression of HLA and costimulatory molecules on macrophages and their phagocytic activity. Finally, MV induces the secretion of inflammatory cytokines, especially IFN-I, and PD-L1 expression in tumor cells and macrophages. These results show that macrophages reduce viral proteins expression in some MPM cell lines through their IFN-I production and generate a pro-inflammatory interplay that may stimulate the patient's anti-tumor immune response.


Assuntos
Técnicas de Cocultura , Macrófagos , Vírus do Sarampo , Terapia Viral Oncolítica , Vírus Oncolíticos , Microambiente Tumoral , Humanos , Vírus do Sarampo/genética , Vírus do Sarampo/fisiologia , Microambiente Tumoral/imunologia , Macrófagos/metabolismo , Macrófagos/imunologia , Macrófagos/virologia , Vírus Oncolíticos/genética , Terapia Viral Oncolítica/métodos , Linhagem Celular Tumoral , Mesotelioma Maligno/patologia , Mesotelioma Maligno/terapia , Interferon Tipo I/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/virologia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/terapia , Neoplasias Pulmonares/virologia , Diferenciação Celular
2.
Microbes Infect ; 26(4): 105321, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38461968

RESUMO

Rabies virus (RABV) is a lethal neurotropic virus that causes 60,000 human deaths every year globally. RABV infection is characterized by the suppression of the interferon (IFN)-mediated antiviral response. However, molecular mechanisms leading to RABV sensing by RIG-I-like receptors (RLR) that initiates IFN signaling currently remain elusive. Here, we showed that RABV RNAs are primarily recognized by the RIG-I RLR, resulting in an IFN response in the infected cells, but this response varied according to the type of RABV used. Pathogenic RABV strain RNAs, Tha, were poorly detected in the cytosol by RIG-I and therefore caused a weak antiviral response. However, we revealed a strong IFN activity triggered by the attenuated RABV vaccine strain RNAs, SAD, mediated by RIG-I. We characterized two major 5' copy-back defective interfering (5'cb DI) genomes generated during SAD replication. Furthermore, we identified an interaction between 5'cb DI genomes, and RIG-I correlated with a high stimulation of the type I IFN signaling. This study indicates that wild-type RABV RNAs poorly activate the RIG-I pathway, while the presence of 5'cb DIs in the live-attenuated vaccine strain serves as an intrinsic adjuvant that strengthens its efficiency by enhancing RIG-I detection thus strongly stimulates the IFN response.


Assuntos
Proteína DEAD-box 58 , Vírus da Raiva , Humanos , Linhagem Celular , Proteína DEAD-box 58/metabolismo , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/imunologia , Interferon Tipo I/metabolismo , Interferon Tipo I/imunologia , Raiva/imunologia , Raiva/virologia , Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Vírus da Raiva/genética , Vírus da Raiva/patogenicidade , Receptores Imunológicos/metabolismo , RNA Viral/genética , Transdução de Sinais , Replicação Viral
3.
Cells ; 13(2)2024 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-38247817

RESUMO

The membrane (M) glycoprotein of coronaviruses (CoVs) serves as the nidus for virion assembly. Using a yeast two-hybrid screen, we identified the interaction of the cytosolic tail of Murine Hepatitis Virus (MHV-CoV) M protein with Myosin Vb (MYO5B), specifically with the alternative splice variant of cellular MYO5B including exon D (MYO5B+D), which mediates interaction with Rab10. When co-expressed in human lung epithelial A549 and canine kidney epithelial MDCK cells, MYO5B+D co-localized with the MHV-CoV M protein, as well as with the M proteins from Porcine Epidemic Diarrhea Virus (PEDV-CoV), Middle East Respiratory Syndrome (MERS-CoV) and Severe Acute Respiratory Syndrome 2 (SARS-CoV-2). Co-expressed M proteins and MYO5B+D co-localized with endogenous Rab10 and Rab11a. We identified point mutations in MHV-CoV M that blocked the interaction with MYO5B+D in yeast 2-hybrid assays. One of these point mutations (E121K) was previously shown to block MHV-CoV virion assembly and its interaction with MYO5B+D. The E to K mutation at homologous positions in PEDV-CoV, MERS-CoV and SARS-CoV-2 M proteins also blocked colocalization with MYO5B+D. The knockdown of Rab10 blocked the co-localization of M proteins with MYO5B+D and was rescued by re-expression of CFP-Rab10. Our results suggest that CoV M proteins traffic through Rab10-containing systems, in association with MYO5B+D.


Assuntos
Proteínas M de Coronavírus , Animais , Cães , Humanos , Células Madin Darby de Rim Canino/metabolismo , Células Madin Darby de Rim Canino/virologia , Coronavírus da Síndrome Respiratória do Oriente Médio , Miosinas , Proteínas rab de Ligação ao GTP/genética , Saccharomyces cerevisiae , Suínos , Proteínas da Matriz Viral , SARS-CoV-2/metabolismo , Vírus da Hepatite Murina/metabolismo , Células A549/metabolismo , Células A549/virologia , Vírus da Diarreia Epidêmica Suína/metabolismo
4.
J Infect Dis ; 228(12): 1675-1679, 2023 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-37562051

RESUMO

The use of soluble recombinant angiotensin-converting enzyme 2 (rACE2) as a decoy capable of blocking SARS-CoV-2 entry into cells has been envisaged as a therapeutic strategy to reduce viral loads in patients with severe COVID-19. We engineered a novel form of rACE2, fused to the Epstein-Barr virus antigen P18F3 (rACE2-P18F3), to reorient a preexisting humoral response toward Epstein-Barr virus against SARS-CoV-2 particles. Recombinant ACE2-P18F3 was able to bind to the SARS-CoV-2 spike protein, neutralize viral entry into cells, and promote the phagocytosis of spheres coated with different spike variants by monocytic cells. The results position rACE2-P18F3 as a promising therapeutic candidate to universally block coronavirus cell entry and clear viral particles.


Assuntos
Enzima de Conversão de Angiotensina 2 , Anticorpos Antivirais , COVID-19 , Herpesvirus Humano 4 , Glicoproteína da Espícula de Coronavírus , Humanos , Enzima de Conversão de Angiotensina 2/metabolismo , Anticorpos Antivirais/imunologia , Herpesvirus Humano 4/imunologia , Peptidil Dipeptidase A/genética , Ligação Proteica , Proteínas Recombinantes/metabolismo , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/imunologia
5.
Cancer Immunol Immunother ; 72(10): 3309-3322, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37466668

RESUMO

Antitumor virotherapy stimulates the antitumor immune response during tumor cell lysis induced by oncolytic viruses (OVs). OV can be modified to express additional transgenes that enhance their therapeutic potential. In this study, we armed the spontaneously oncolytic Schwarz strain of measles viruses (MVs) with the gene encoding the cancer/testis antigen NY-ESO-1 to obtain MVny. We compared MV and MVny oncolytic activity and ability to induce NY-ESO-1 expression in six human melanoma cell lines. After MVny infection, we measured the capacity of melanoma cells to present NY-ESO-1 peptides to CD4 + and CD8 + T cell clones specific for this antigen. We assessed the ability of MVny to induce NY-ESO-1 expression and presentation in monocyte-derived dendritic cells (DCs). Our results show that MVny and MV oncolytic activity are similar with a faster cell lysis induced by MVny. We also observed that melanoma cell lines and DC expressed the NY-ESO-1 protein after MVny infection. In addition, MVny-infected melanoma cells and DCs were able to stimulate NY-ESO-1-specific CD4 + and CD8 + T cells. Finally, MVny was able to induce DC maturation. Altogether, these results show that MVny could be an interesting candidate to stimulate NY-ESO-1-specific T cells in melanoma patients with NY-ESO-1-expressing tumor cells.


Assuntos
Sarampo , Melanoma , Vírus Oncolíticos , Masculino , Humanos , Vírus Oncolíticos/genética , Proteínas de Membrana , Vírus do Sarampo/genética , Melanoma/metabolismo , Linfócitos T CD8-Positivos , Antígenos de Neoplasias , Anticorpos/metabolismo , Células Dendríticas , Sarampo/metabolismo
6.
Med Sci (Paris) ; 38(12): 1052-1060, 2022 Dec.
Artigo em Francês | MEDLINE | ID: mdl-36692265

RESUMO

Viruses have been used as tools to prevent viral infections themselves for more than two centuries with impressive success. After the empirical discoveries of the first vaccines, today the development of genetic engineering, molecular virology, reverse genetics, the manipulation of viral genomes, their high-throughput sequencing and their chemical synthesis, the mastery of cell culture and purification methods have greatly benefited the development of viral vaccines. Since smallpox and rabies, the history of vaccinology has followed in the footsteps of the history of virology. New mRNA or viral vector vaccines have emerged in recent years. They were developed and distributed to the population in record time in the face of the Covid pandemic. Viruses in the service of health have a bright future ahead of them, whether to prevent other pandemics, to treat cancer, or to finally control HIV and malaria.


Title: Les virus au service de la santé : la vaccination. Abstract: Depuis plus de deux siècles, les virus sont utilisés, avec un succès impressionnant, comme outils de prévention des infections virales. Depuis la variole et la rage, l'histoire de la vaccinologie a suivi les pas de l'histoire de la virologie. Après les découvertes empiriques des premiers vaccins, le développement du génie génétique, de la virologie moléculaire, de la génétique inverse, la manipulation des génomes viraux, leur séquençage à haut débit et leur synthèse chimique, la maîtrise de la culture cellulaire et des méthodes de purification, ont considérablement contribué au développement de nouveaux vaccins viraux. Des vaccins à ARN messager ou à vecteur viral ont ainsi vu le jour ces dernières années et, face à la pandémie de Covid-19, ont été développés et distribués à la population en un temps record. Les virus au service de la santé ont un bel avenir devant eux, que cela soit pour prévenir d'autres pandémies, pour traiter le cancer, ou contrôler, enfin, le VIH ou le Plasmodium, l'agent du paludisme.


Assuntos
COVID-19 , Vacinas Virais , Viroses , Vírus , Humanos , COVID-19/prevenção & controle , Vacinação/história , Viroses/prevenção & controle
7.
Nat Commun ; 12(1): 6277, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34725327

RESUMO

Several COVID-19 vaccines have now been deployed to tackle the SARS-CoV-2 pandemic, most of them based on messenger RNA or adenovirus vectors.The duration of protection afforded by these vaccines is unknown, as well as their capacity to protect from emerging new variants. To provide sufficient coverage for the world population, additional strategies need to be tested. The live pediatric measles vaccine (MV) is an attractive approach, given its extensive safety and efficacy history, along with its established large-scale manufacturing capacity. We develop an MV-based SARS-CoV-2 vaccine expressing the prefusion-stabilized, membrane-anchored full-length S antigen, which proves to be efficient at eliciting strong Th1-dominant T-cell responses and high neutralizing antibody titers. In both mouse and golden Syrian hamster models, these responses protect the animals from intranasal infectious challenge. Additionally, the elicited antibodies efficiently neutralize in vitro the three currently circulating variants of SARS-CoV-2.


Assuntos
Vacinas contra COVID-19/imunologia , COVID-19/imunologia , Vetores Genéticos , Imunidade , Adenoviridae , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , COVID-19/prevenção & controle , Vacinas contra COVID-19/administração & dosagem , Cricetinae , Citocinas , Feminino , Imunização , Imunização Secundária , Masculino , Vacina contra Sarampo/imunologia , Mesocricetus , Camundongos , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/imunologia
8.
J Virol ; 95(22): e0091221, 2021 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-34431698

RESUMO

Respiratory syncytial virus (RSV) is the main cause of acute respiratory infections in young children and also has a major impact on the elderly and immunocompromised people. In the absence of a vaccine or efficient treatment, a better understanding of RSV interactions with the host antiviral response during infection is needed. Previous studies revealed that cytoplasmic inclusion bodies (IBs), where viral replication and transcription occur, could play a major role in the control of innate immunity during infection by recruiting cellular proteins involved in the host antiviral response. We recently showed that the morphogenesis of IBs relies on a liquid-liquid-phase separation mechanism depending on the interaction between viral nucleoprotein (N) and phosphoprotein (P). These scaffold proteins are expected to play a central role in the recruitment of cellular proteins to IBs. Here, we performed a yeast two-hybrid screen using RSV N protein as bait and identified the cellular protein TAX1BP1 as a potential partner of this viral protein. This interaction was validated by pulldown and immunoprecipitation assays. We showed that TAX1BP1 suppression has only a limited impact on RSV infection in cell cultures. However, RSV replication is decreased in TAX1BP1-deficient (TAX1BP1 knockout [TAX1BP1KO]) mice, whereas the production of inflammatory and antiviral cytokines is enhanced. In vitro infection of wild-type or TAX1BP1KO alveolar macrophages confirmed that the innate immune response to RSV infection is enhanced in the absence of TAX1BP1. Altogether, our results suggest that RSV could hijack TAX1BP1 to restrain the host immune response during infection. IMPORTANCE Respiratory syncytial virus (RSV), which is the leading cause of lower respiratory tract illness in infants, remains a medical problem in the absence of a vaccine or efficient treatment. This virus is also recognized as a main pathogen in the elderly and immunocompromised people, and the occurrence of coinfections (with other respiratory viruses and bacteria) amplifies the risks of developing respiratory distress. In this context, a better understanding of the pathogenesis associated with viral respiratory infections, which depends on both viral replication and the host immune response, is needed. The present study reveals that the cellular protein TAX1BP1, which interacts with the RSV nucleoprotein N, participates in the control of the innate immune response during RSV infection, suggesting that the N-TAX1BP1 interaction represents a new target for the development of antivirals.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Proteínas de Neoplasias/imunologia , Proteínas do Nucleocapsídeo/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Animais , Linhagem Celular , Cricetinae , Humanos , Imunidade Inata , Camundongos , Camundongos Knockout , Replicação Viral
9.
Mol Cell Proteomics ; 20: 100049, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33515806

RESUMO

Viruses manipulate the central machineries of host cells to their advantage. They prevent host cell antiviral responses to create a favorable environment for their survival and propagation. Measles virus (MV) encodes two nonstructural proteins MV-V and MV-C known to counteract the host interferon response and to regulate cell death pathways. Several molecular mechanisms underlining MV-V regulation of innate immunity and cell death pathways have been proposed, whereas MV-C host-interacting proteins are less studied. We suggest that some cellular factors that are controlled by MV-C protein during viral replication could be components of innate immunity and the cell death pathways. To determine which host factors are targeted by MV-C, we captured both direct and indirect host-interacting proteins of MV-C protein. For this, we used a strategy based on recombinant viruses expressing tagged viral proteins followed by affinity purification and a bottom-up mass spectrometry analysis. From the list of host proteins specifically interacting with MV-C protein in different cell lines, we selected the host targets that belong to immunity and cell death pathways for further validation. Direct protein interaction partners of MV-C were determined by applying protein complementation assay and the bioluminescence resonance energy transfer approach. As a result, we found that MV-C protein specifically interacts with p65-iASPP protein complex that controls both cell death and innate immunity pathways and evaluated the significance of these host factors on virus replication.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Repressoras/metabolismo , Fator de Transcrição RelA/metabolismo , Proteínas não Estruturais Virais/metabolismo , Animais , Morte Celular , Linhagem Celular , Chlorocebus aethiops , Interações Hospedeiro-Patógeno , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Vírus do Sarampo/genética , Vírus do Sarampo/fisiologia , Mapas de Interação de Proteínas , Proteômica , Proteínas Repressoras/genética , Fator de Transcrição RelA/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteínas não Estruturais Virais/genética , Replicação Viral
10.
J Virol ; 94(22)2020 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-32878892

RESUMO

Yellow fever virus (YFV) is an RNA virus primarily targeting the liver. Severe YF cases are responsible for hemorrhagic fever, plausibly precipitated by excessive proinflammatory cytokine response. Pathogen recognition receptors (PRRs), such as the cytoplasmic retinoic acid inducible gene I (RIG-I)-like receptors (RLRs), and the viral RNA sensor protein kinase R (PKR), are known to initiate a proinflammatory response upon recognition of viral genomes. Here, we sought to reveal the main determinants responsible for the acute cytokine expression occurring in human hepatocytes following YFV infection. Using a RIG-I-defective human hepatoma cell line, we found that RIG-I largely contributes to cytokine secretion upon YFV infection. In infected RIG-I-proficient hepatoma cells, RIG-I was localized in stress granules. These granules are large aggregates of stalled translation preinitiation complexes known to concentrate RLRs and PKR and are so far recognized as hubs orchestrating RNA virus sensing. Stable knockdown of PKR in hepatoma cells revealed that PKR contributes to both stress granule formation and cytokine induction upon YFV infection. However, stress granule disruption did not affect the cytokine response to YFV infection, as assessed by small interfering RNA (siRNA)-knockdown-mediated inhibition of stress granule assembly. Finally, no viral RNA was detected in stress granules using a fluorescence in situ hybridization approach coupled with immunofluorescence. Our findings suggest that both RIG-I and PKR mediate proinflammatory cytokine induction in YFV-infected hepatocytes, in a stress granule-independent manner. Therefore, by showing the uncoupling of the cytokine response from the stress granule formation, our model challenges the current view in which stress granules are required for the mounting of the acute antiviral response.IMPORTANCE Yellow fever is a mosquito-borne acute hemorrhagic disease caused by yellow fever virus (YFV). The mechanisms responsible for its pathogenesis remain largely unknown, although increased inflammation has been linked to worsened outcome. YFV targets the liver, where it primarily infects hepatocytes. We found that two RNA-sensing proteins, RIG-I and PKR, participate in the induction of proinflammatory mediators in human hepatocytes infected with YFV. We show that YFV infection promotes the formation of cytoplasmic structures, termed stress granules, in a PKR- but not RIG-I-dependent manner. While stress granules were previously postulated to be essential platforms for immune activation, we found that they are not required for the production of proinflammatory mediators upon YFV infection. Collectively, our work uncovered molecular events triggered by the replication of YFV, which could prove instrumental in clarifying the pathogenesis of the disease, with possible repercussions for disease management.


Assuntos
Proteína DEAD-box 58/metabolismo , Vírus da Febre Amarela/metabolismo , eIF-2 Quinase/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Carcinoma Hepatocelular , Linhagem Celular , Linhagem Celular Tumoral , Citocinas/metabolismo , Proteína DEAD-box 58/deficiência , Proteína DEAD-box 58/genética , DNA Helicases/genética , Técnicas de Silenciamento de Genes , Haplorrinos , Hepatócitos/virologia , Humanos , Proteínas de Ligação a Poli-ADP-Ribose/genética , RNA Helicases/genética , Proteínas com Motivo de Reconhecimento de RNA/genética , RNA Interferente Pequeno , RNA Viral/genética , Proteínas de Ligação a RNA/genética , Receptores Imunológicos , Antígeno-1 Intracelular de Células T/genética , Transcriptoma , eIF-2 Quinase/genética
12.
J Thorac Oncol ; 15(5): 827-842, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31945495

RESUMO

INTRODUCTION: Oncolytic immunotherapy is based on the use of nonpathogenic replicative oncolytic viruses that infect and kill tumor cells exclusively. Recently, we found that the spontaneous oncolytic activity of the Schwarz strain of measles virus (MV) against human malignant pleural mesothelioma (MPM) depends on defects in the antiviral type I interferon (IFN-I) response in tumor cells. METHODS: In this study, we studied three independent human MPM bio-collections to identify the defects in the IFN-I responses in tumor cells. RESULTS: We show that the most frequent defect is the homozygous deletions (HDs) of all the 14 IFN-I genes (IFN-α and IFN-ß) that we found in more than half of MV-sensitive MPM cell lines. These HDs occur together with the HDs of the tumor suppressor gene CDKN2A also located in the 9p21.3 chromosome region. Therefore, the IFN-I-/- MPM cell lines develop a partial and weak IFN-I response when they are exposed to the virus compared with that of normal cells and MV-resistant MPM cell lines. This response consists of the expression of a restricted number of IFN-stimulated genes that do not depend on the presence of IFN-I. In addition, the IFN-I-/- MPM cell lines infected by MV also develop a pro-inflammatory response associated with stress of the endoplasmic reticulum. CONCLUSION: Our study emphasizes the link between HDs of IFN-I encoding genes and the CDKN2A gene in MPM and sensitivity to MV oncolytic immunotherapy.


Assuntos
Interferon Tipo I , Neoplasias Pulmonares , Mesotelioma , Terapia Viral Oncolítica , Vírus Oncolíticos , Linhagem Celular Tumoral , Homozigoto , Humanos , Interferon Tipo I/genética , Vírus do Sarampo/genética , Mesotelioma/genética , Mesotelioma/terapia , Vírus Oncolíticos/genética , Deleção de Sequência
13.
Methods Mol Biol ; 2058: 127-132, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31486035

RESUMO

Oncolytic immunotherapy efficacy relies partially on the induction of immunogenic tumor cell death following infection with oncolytic viruses (OV) to induce an antitumor immune response. Here, we describe a method to determine if an OV is able to induce such an immunogenic tumor cell death. This method consists in testing whether tumor cells lysed by an OV are able to induce the maturation of human monocyte-derived immature dendritic cells (Mo-iDC).


Assuntos
Vetores Genéticos/genética , Imunomodulação , Neoplasias/imunologia , Vírus Oncolíticos/genética , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Morte Celular/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Terapia Genética/métodos , Humanos , Imunofenotipagem , Monócitos/imunologia , Monócitos/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/imunologia
14.
Sci Rep ; 9(1): 20134, 2019 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-31882812

RESUMO

T. capitatus is widely used in traditional medicine in Tunisia. The main goal of this study was to evaluate the phytochemical composition, the fatty acids profile, the antioxidant, antibacterial, and antifungal activities as well as the cytotoxic potential of the essential oil (EO) of this plant. The identification and the quantification of the different constituents of the tested EO was determined by gas chromatography-mass spectrometry (GC-MS). Antioxidant activities were evaluated by spectrophotometric methods and chemical tests. HCT 116 cells were used to evaluate the cytotoxic effect of the EO. The microdilution method was conducted to evaluate the antibacterial activity. Poisoned food method was used to test the antifungal activities against fungi species such Aspergillus niger and Aspergillus flavus. The EO presented several components, mainly monoterpenes. Results revealed that T. capitatus EO is not cytotoxic and showed excellent antioxidant activity with a dose dependent manner. Regarding antimicrobial activity, T. capitatus EO was efficient against all tested bacteria and fungi.


Assuntos
Ácidos Graxos/química , Óleos Voláteis/química , Óleos Voláteis/farmacologia , Compostos Fitoquímicos/química , Extratos Vegetais/química , Thymus (Planta)/química , Anti-Infecciosos/química , Anti-Infecciosos/farmacologia , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacologia , Antioxidantes/química , Antioxidantes/farmacologia , Bactérias/efeitos dos fármacos , Linhagem Celular , Relação Dose-Resposta a Droga , Fungos/efeitos dos fármacos , Humanos , Testes de Sensibilidade Microbiana , Óleos Voláteis/análise , Compostos Fitoquímicos/farmacologia , Extratos Vegetais/farmacologia
15.
Cell Host Microbe ; 26(6): 823-835.e11, 2019 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-31761719

RESUMO

RNA-binding proteins (RBPs) perform key cellular activities by controlling the function of bound RNAs. The widely held assumption that RBPs are strictly intracellular has been challenged by the discovery of secreted RBPs. However, extracellular RBPs have been described in eukaryotes, while secreted bacterial RBPs have not been reported. Here, we show that the bacterial pathogen Listeria monocytogenes secretes a small RBP that we named Zea. We show that Zea binds a subset of L. monocytogenes RNAs, causing their accumulation in the extracellular medium. Furthermore, during L. monocytogenes infection, Zea binds RIG-I, the non-self-RNA innate immunity sensor, potentiating interferon-ß production. Mouse infection studies reveal that Zea affects L. monocytogenes virulence. Together, our results unveil that bacterial RNAs can be present extracellularly in association with RBPs, acting as "social RNAs" to trigger a host response during infection.


Assuntos
Proteína DEAD-box 58/metabolismo , Listeria monocytogenes/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Proteína DEAD-box 58/imunologia , Células HEK293 , Interações entre Hospedeiro e Microrganismos , Humanos , Imunidade Inata , Interferon beta/metabolismo , Listeria monocytogenes/imunologia , Listeria monocytogenes/patogenicidade , Camundongos , RNA Bacteriano/metabolismo , Transdução de Sinais/imunologia , Virulência/imunologia
16.
Sci Signal ; 12(601)2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31575732

RESUMO

The retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) RIG-I, MDA5, and LGP2 stimulate inflammatory and antiviral responses by sensing nonself RNA molecules produced during viral replication. Here, we investigated how LGP2 regulates the RIG-I- and MDA5-dependent induction of type I interferon (IFN) signaling and showed that LGP2 interacted with different components of the RNA-silencing machinery. We identified a direct protein-protein interaction between LGP2 and the IFN-inducible, double-stranded RNA binding protein PACT. The LGP2-PACT interaction was mediated by the regulatory C-terminal domain of LGP2 and was necessary for inhibiting RIG-I-dependent responses and for amplifying MDA5-dependent responses. We described a point mutation within LGP2 that disrupted the LGP2-PACT interaction and led to the loss of LGP2-mediated regulation of RIG-I and MDA5 signaling. These results suggest a model in which the LGP2-PACT interaction regulates the inflammatory responses mediated by RIG-I and MDA5 and enables the cellular RNA-silencing machinery to coordinate with the innate immune response.


Assuntos
Antivirais/metabolismo , Proteína DEAD-box 58/metabolismo , Helicase IFIH1 Induzida por Interferon/metabolismo , RNA Helicases/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Chlorocebus aethiops , Proteína DEAD-box 58/genética , Enterovirus Humano B/genética , Enterovirus Humano B/fisiologia , Células HEK293 , Células HeLa , Humanos , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Helicase IFIH1 Induzida por Interferon/genética , Mengovirus/genética , Mengovirus/fisiologia , Ligação Proteica , RNA Helicases/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Proteínas de Ligação a RNA/genética , Receptores Imunológicos , Transdução de Sinais/genética , Células Vero
17.
Viruses ; 11(3)2019 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-30909570

RESUMO

Lassa virus (LASV) and Mopeia virus (MOPV) are two closely related Old-World mammarenaviruses. LASV causes severe hemorrhagic fever with high mortality in humans, whereas no case of MOPV infection has been reported. Comparing MOPV and LASV is a powerful strategy to unravel pathogenic mechanisms that occur during the course of pathogenic arenavirus infection. We used a yeast two-hybrid approach to identify cell partners of MOPV and LASV Z matrix protein in which two autophagy adaptors were identified, NDP52 and TAX1BP1. Autophagy has emerged as an important cellular defense mechanism against viral infections but its role during arenavirus infection has not been shown. Here, we demonstrate that autophagy is transiently induced by MOPV, but not LASV, in infected cells two days after infection. Impairment of the early steps of autophagy significantly decreased the production of MOPV and LASV infectious particles, whereas a blockade of the degradative steps impaired only MOPV infectious particle production. Our study provides insights into the role played by autophagy during MOPV and LASV infection and suggests that this process could partially explain their different pathogenicity.


Assuntos
Arenavirus/fisiologia , Autofagia , Vírus Lassa/fisiologia , Animais , Arenavirus/patogenicidade , Chlorocebus aethiops , Células HEK293 , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Vírus Lassa/patogenicidade , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Técnicas do Sistema de Duplo-Híbrido , Células Vero
18.
Front Immunol ; 10: 134, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30809223

RESUMO

The guanabenz derivative Sephin1 has recently been proposed to increase the levels of translation initiation factor 2 (eIF2α) phosphorylation by inhibiting dephosphorylation by the protein phosphatase 1-GADD34 (PPP1R15A) complex. As phosphorylation of eIF2α by protein kinase R (PKR) is a prominent cellular antiviral pathway, we evaluated the consequences of Sephin1 treatment on virus replication. Our results provide evidence that Sephin1 downregulates replication of human respiratory syncytial virus, measles virus, human adenovirus 5 virus, human enterovirus D68, human cytomegalovirus, and rabbit myxoma virus. However, Sephin1 proved to be inactive against influenza virus, as well as against Japanese encephalitis virus. Sephin1 increased the levels of phosphorylated eIF2α in cells exposed to a PKR agonist. By contrast, in virus-infected cells, the levels of phosphorylated eIF2α did not always correlate with the inhibition of virus replication by Sephin1. This work identifies Sephin1 as an antiviral molecule in cell culture against RNA, as well as DNA viruses belonging to phylogenetically distant families.


Assuntos
Antivirais/farmacologia , Fator de Iniciação 2 em Eucariotos/metabolismo , Guanabenzo/análogos & derivados , Animais , Antivirais/uso terapêutico , Linhagem Celular , Vírus de DNA/efeitos dos fármacos , Vírus de DNA/fisiologia , Guanabenzo/farmacologia , Guanabenzo/uso terapêutico , Humanos , Camundongos , Fosforilação/efeitos dos fármacos , Infecções por Poxviridae/tratamento farmacológico , Vírus de RNA/efeitos dos fármacos , Vírus de RNA/fisiologia , Coelhos , Infecções Tumorais por Vírus/tratamento farmacológico , Replicação Viral/efeitos dos fármacos
19.
Cancer Immunol Immunother ; 68(4): 533-544, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30656384

RESUMO

Cancer immunotherapy is seeing an increasing focus on vaccination with tumor-associated antigens (TAAs). Human telomerase (hTERT) is a TAA expressed by most tumors to overcome telomere shortening. Tolerance to hTERT can be easily broken both naturally and experimentally and hTERT DNA vaccine candidates have been introduced in clinical trials. DNA prime/boost strategies have been widely developed to immunize efficiently against infectious diseases. We explored the use of a recombinant measles virus (MV) hTERT vector to boost DNA priming as recombinant live attenuated measles virus has an impressive safety and efficacy record. Here, we show that a MV-TERT vector can rapidly and strongly boost DNA hTERT priming in MV susceptible IFNAR/CD46 mouse models. The cellular immune responses were Th1 polarized. No humoral responses were elicited. The 4 kb hTERT transgene did not impact MV replication or induction of cell-mediated responses. These findings validate the MV-TERT vector to boost cell-mediated responses following DNA priming in humans.


Assuntos
Vacinas Anticâncer/imunologia , Epitopos de Linfócito T/imunologia , Vetores Genéticos , Imunidade Celular , Vírus do Sarampo , Linfócitos T/imunologia , Telomerase/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Vacinas Anticâncer/genética , Linhagem Celular , Chlorocebus aethiops , Citocinas/metabolismo , Citotoxicidade Imunológica , Vetores Genéticos/genética , Humanos , Imunização , Imunização Secundária , Vírus do Sarampo/genética , Camundongos , Camundongos Transgênicos , Telomerase/genética , Vacinas de DNA , Células Vero
20.
Blood Adv ; 2(23): 3492-3505, 2018 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-30530776

RESUMO

In this study, we assessed the sensitivity of myeloma cells to the oncolytic measles virus (MV) in relation to p53 using 37 cell lines and 23 primary samples. We showed that infection and cell death were correlated with CD46 expression, which was associated with TP53 status; TP53 abn cell lines highly expressed CD46 and were preferentially infected by MV when compared with the TP53 wt cell lines (P = .046 and P = .045, respectively). Infection of myeloma cells was fully dependent on CD46 expression in both cell lines and primary cells. In the TP53 wt cell lines, but not the TP53 abn cell lines, activation of the p53 pathway with nutlin3a inhibited both CD46 expression and MV infection, while TP53 silencing reciprocally increased CD46 expression and MV infection. We showed using a p53 chromatin immunoprecipitation assay and microRNA assessment that CD46 gene expression was directly and indirectly regulated by p53. Primary myeloma cells overexpressed CD46 as compared with normal cells and were highly infected and killed by MV. CD46 expression and MV infection were inhibited by nutlin3a in primary p53-competent myeloma cells, but not in p53-deficient myeloma cells, and the latter were highly sensitive to MV infection. In summary, myeloma cells were highly sensitive to MV and infection inhibition by the p53 pathway was abrogated in p53-deficient myeloma cells. These results argue for an MV-based clinical trial for patients with p53 deficiency.


Assuntos
Vírus do Sarampo/fisiologia , Proteína Cofatora de Membrana/metabolismo , Mieloma Múltiplo/patologia , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Humanos , Proteína Cofatora de Membrana/antagonistas & inibidores , Proteína Cofatora de Membrana/genética , MicroRNAs/metabolismo , Mieloma Múltiplo/metabolismo , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/química , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA