Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.677
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 182(2): 532-532.e1, 2020 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-32707094

RESUMO

Influenza is one of the best-studied viruses of all time, and as such, it serves as a testbed to extend our biological knowledge to the nanoscale. Many of the key processes underlying influenza infection and our antibody response against the virus have been thoroughly investigated. This SnapShot describes these key numbers for prototypical lab-adapted strains of the human influenza A virus. To view this SnapShot, open or download the PDF.


Assuntos
Vírus da Influenza A/metabolismo , Influenza Humana/patologia , Formação de Anticorpos , Eritrócitos/virologia , Hemaglutininas/química , Hemaglutininas/metabolismo , Humanos , Vírus da Influenza A/imunologia , Vírus da Influenza A/patogenicidade , Influenza Humana/virologia , Neuraminidase/química , Neuraminidase/metabolismo , Ligação Proteica , Estrutura Quaternária de Proteína , Ácidos Siálicos/metabolismo
2.
Nat Immunol ; 22(4): 497-509, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33790474

RESUMO

Classic major histocompatibility complex class I (MHC-I) presentation relies on shuttling cytosolic peptides into the endoplasmic reticulum (ER) by the transporter associated with antigen processing (TAP). Viruses disable TAP to block MHC-I presentation and evade cytotoxic CD8+ T cells. Priming CD8+ T cells against these viruses is thought to rely solely on cross-presentation by uninfected TAP-functional dendritic cells. We found that protective CD8+ T cells could be mobilized during viral infection even when TAP was absent in all hematopoietic cells. TAP blockade depleted the endosomal recycling compartment of MHC-I molecules and, as such, impaired Toll-like receptor-regulated cross-presentation. Instead, MHC-I molecules accumulated in the ER-Golgi intermediate compartment (ERGIC), sequestered away from Toll-like receptor control, and coopted ER-SNARE Sec22b-mediated vesicular traffic to intersect with internalized antigen and rescue cross-presentation. Thus, when classic MHC-I presentation and endosomal recycling compartment-dependent cross-presentation are impaired in dendritic cells, cell-autonomous noncanonical cross-presentation relying on ERGIC-derived MHC-I counters TAP dysfunction to nevertheless mediate CD8+ T cell priming.


Assuntos
Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Linfócitos T CD8-Positivos/imunologia , Apresentação Cruzada , Células Dendríticas/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Vírus da Influenza A/imunologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Modelos Animais de Doenças , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/virologia , Feminino , Complexo de Golgi/imunologia , Complexo de Golgi/metabolismo , Complexo de Golgi/virologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza A/patogenicidade , Ativação Linfocitária , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Orthomyxoviridae/genética
3.
Cell ; 172(6): 1157-1159, 2018 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-29522735

RESUMO

100 years after the infamous "Spanish flu" pandemic, the 2017-2018 flu season has been severe, with numerous infections worldwide. In between, there have been continuous, relentless attacks from (re-)emerging viruses. To fully understand viral pathogenesis and develop effective medical countermeasures, we must strengthen current surveillance and basic research efforts.


Assuntos
Vírus da Influenza A Subtipo H5N2/patogenicidade , Influenza Aviária/virologia , Infecção por Zika virus/virologia , Zika virus/patogenicidade , Animais , Aves , Doenças Transmissíveis Emergentes/epidemiologia , Doenças Transmissíveis Emergentes/virologia , Humanos , Vírus da Influenza A/classificação , Vírus da Influenza A/patogenicidade , Influenza Aviária/epidemiologia , Influenza Humana/epidemiologia , Influenza Humana/virologia , Pandemias , Filogeografia , Infecção por Zika virus/epidemiologia
4.
Nature ; 628(8009): 835-843, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38600381

RESUMO

Severe influenza A virus (IAV) infections can result in hyper-inflammation, lung injury and acute respiratory distress syndrome1-5 (ARDS), for which there are no effective pharmacological therapies. Necroptosis is an attractive entry point for therapeutic intervention in ARDS and related inflammatory conditions because it drives pathogenic lung inflammation and lethality during severe IAV infection6-8 and can potentially be targeted by receptor interacting protein kinase 3 (RIPK3) inhibitors. Here we show that a newly developed RIPK3 inhibitor, UH15-38, potently and selectively blocked IAV-triggered necroptosis in alveolar epithelial cells in vivo. UH15-38 ameliorated lung inflammation and prevented mortality following infection with laboratory-adapted and pandemic strains of IAV, without compromising antiviral adaptive immune responses or impeding viral clearance. UH15-38 displayed robust therapeutic efficacy even when administered late in the course of infection, suggesting that RIPK3 blockade may provide clinical benefit in patients with IAV-driven ARDS and other hyper-inflammatory pathologies.


Assuntos
Lesão Pulmonar , Necroptose , Infecções por Orthomyxoviridae , Inibidores de Proteínas Quinases , Proteína Serina-Treonina Quinases de Interação com Receptores , Animais , Feminino , Humanos , Masculino , Camundongos , Células Epiteliais Alveolares/patologia , Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/virologia , Células Epiteliais Alveolares/metabolismo , Vírus da Influenza A/classificação , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/imunologia , Vírus da Influenza A/patogenicidade , Lesão Pulmonar/complicações , Lesão Pulmonar/patologia , Lesão Pulmonar/prevenção & controle , Lesão Pulmonar/virologia , Camundongos Endogâmicos C57BL , Necroptose/efeitos dos fármacos , Infecções por Orthomyxoviridae/complicações , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/mortalidade , Infecções por Orthomyxoviridae/virologia , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/antagonistas & inibidores , Síndrome do Desconforto Respiratório/complicações , Síndrome do Desconforto Respiratório/patologia , Síndrome do Desconforto Respiratório/prevenção & controle , Síndrome do Desconforto Respiratório/virologia
5.
Nature ; 621(7977): 120-128, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37558883

RESUMO

Humans display substantial interindividual clinical variability after SARS-CoV-2 infection1-3, the genetic and immunological basis of which has begun to be deciphered4. However, the extent and drivers of population differences in immune responses to SARS-CoV-2 remain unclear. Here we report single-cell RNA-sequencing data for peripheral blood mononuclear cells-from 222 healthy donors of diverse ancestries-that were stimulated with SARS-CoV-2 or influenza A virus. We show that SARS-CoV-2 induces weaker, but more heterogeneous, interferon-stimulated gene activity compared with influenza A virus, and a unique pro-inflammatory signature in myeloid cells. Transcriptional responses to viruses display marked population differences, primarily driven by changes in cell abundance including increased lymphoid differentiation associated with latent cytomegalovirus infection. Expression quantitative trait loci and mediation analyses reveal a broad effect of cell composition on population disparities in immune responses, with genetic variants exerting a strong effect on specific loci. Furthermore, we show that natural selection has increased population differences in immune responses, particularly for variants associated with SARS-CoV-2 response in East Asians, and document the cellular and molecular mechanisms by which Neanderthal introgression has altered immune functions, such as the response of myeloid cells to viruses. Finally, colocalization and transcriptome-wide association analyses reveal an overlap between the genetic basis of immune responses to SARS-CoV-2 and COVID-19 severity, providing insights into the factors contributing to current disparities in COVID-19 risk.


Assuntos
COVID-19 , Genética Populacional , SARS-CoV-2 , Análise da Expressão Gênica de Célula Única , Animais , Humanos , Diferenciação Celular , COVID-19/genética , COVID-19/imunologia , COVID-19/virologia , Citomegalovirus/fisiologia , População do Leste Asiático/genética , Introgressão Genética , Vírus da Influenza A/patogenicidade , Vírus da Influenza A/fisiologia , Interferons/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Células Mieloides/imunologia , Homem de Neandertal/genética , Homem de Neandertal/imunologia , SARS-CoV-2/genética , SARS-CoV-2/imunologia , SARS-CoV-2/patogenicidade , SARS-CoV-2/fisiologia , Seleção Genética , Latência Viral
6.
Mol Cell ; 81(9): 2031-2040.e8, 2021 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-33909989

RESUMO

Autophagy is a fundamental catabolic process that uses a unique post-translational modification, the conjugation of ATG8 protein to phosphatidylethanolamine (PE). ATG8 lipidation also occurs during non-canonical autophagy, a parallel pathway involving conjugation of ATG8 to single membranes (CASM) at endolysosomal compartments, with key functions in immunity, vision, and neurobiology. It is widely assumed that CASM involves the same conjugation of ATG8 to PE, but this has not been formally tested. Here, we discover that all ATG8s can also undergo alternative lipidation to phosphatidylserine (PS) during CASM, induced pharmacologically, by LC3-associated phagocytosis or influenza A virus infection, in mammalian cells. Importantly, ATG8-PS and ATG8-PE adducts are differentially delipidated by the ATG4 family and bear different cellular dynamics, indicating significant molecular distinctions. These results provide important insights into autophagy signaling, revealing an alternative form of the hallmark ATG8 lipidation event. Furthermore, ATG8-PS provides a specific "molecular signature" for the non-canonical autophagy pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Autofagossomos/metabolismo , Família da Proteína 8 Relacionada à Autofagia/metabolismo , Autofagia , Proteínas Associadas aos Microtúbulos/metabolismo , Fosfatidilserinas/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Autofagossomos/efeitos dos fármacos , Autofagossomos/genética , Autofagossomos/patologia , Família da Proteína 8 Relacionada à Autofagia/genética , Proteínas Relacionadas à Autofagia/genética , Proteínas Relacionadas à Autofagia/metabolismo , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , Feminino , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Vírus da Influenza A/patogenicidade , Macrolídeos/farmacologia , Masculino , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Monensin/farmacologia , Fagocitose , Fosfatidiletanolaminas/metabolismo , Células RAW 264.7 , Transdução de Sinais
7.
Mol Cell ; 73(6): 1162-1173.e5, 2019 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-30712990

RESUMO

The MHC class I antigen presentation system enables T cell immunosurveillance of cancers and viruses. A substantial fraction of the immunopeptidome derives from rapidly degraded nascent polypeptides (DRiPs). By knocking down each of the 80 ribosomal proteins, we identified proteins that modulate peptide generation without altering source protein expression. We show that 60S ribosomal proteins L6 (RPL6) and RPL28, which are adjacent on the ribosome, play opposite roles in generating an influenza A virus-encoded peptide. Depleting RPL6 decreases ubiquitin-dependent peptide presentation, whereas depleting RPL28 increases ubiquitin-dependent and -independent peptide presentation. 40S ribosomal protein S28 (RPS28) knockdown increases total peptide supply in uninfected cells by increasing DRiP synthesis from non-canonical translation of "untranslated" regions and non-AUG start codons and sensitizes tumor cells for T cell targeting. Our findings raise the possibility of modulating immunosurveillance by pharmaceutical targeting ribosomes.


Assuntos
Apresentação de Antígeno , Antígenos de Histocompatibilidade Classe I/biossíntese , Proteínas Ribossômicas/metabolismo , Subunidades Ribossômicas Maiores de Eucariotos/metabolismo , Subunidades Ribossômicas Menores de Eucariotos/metabolismo , Linfócitos T/metabolismo , Animais , Linhagem Celular Tumoral , Técnicas de Cocultura , Células HEK293 , Antígenos de Histocompatibilidade Classe I/imunologia , Interações Hospedeiro-Patógeno , Humanos , Vigilância Imunológica , Vírus da Influenza A/imunologia , Vírus da Influenza A/patogenicidade , Melanoma/imunologia , Melanoma/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Ribossômicas/genética , Subunidades Ribossômicas Maiores de Eucariotos/genética , Subunidades Ribossômicas Menores de Eucariotos/genética , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/metabolismo , Linfócitos T/imunologia , Linfócitos T/virologia
8.
Nature ; 587(7834): 466-471, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33116313

RESUMO

Severe respiratory infections can result in acute respiratory distress syndrome (ARDS)1. There are no effective pharmacological therapies that have been shown to improve outcomes for patients with ARDS. Although the host inflammatory response limits spread of and eventually clears the pathogen, immunopathology is a major contributor to tissue damage and ARDS1,2. Here we demonstrate that respiratory viral infection induces distinct fibroblast activation states, which we term extracellular matrix (ECM)-synthesizing, damage-responsive and interferon-responsive states. We provide evidence that excess activity of damage-responsive lung fibroblasts drives lethal immunopathology during severe influenza virus infection. By producing ECM-remodelling enzymes-in particular the ECM protease ADAMTS4-and inflammatory cytokines, damage-responsive fibroblasts modify the lung microenvironment to promote robust immune cell infiltration at the expense of lung function. In three cohorts of human participants, the levels of ADAMTS4 in the lower respiratory tract were associated with the severity of infection with seasonal or avian influenza virus. A therapeutic agent that targets the ECM protease activity of damage-responsive lung fibroblasts could provide a promising approach to preserving lung function and improving clinical outcomes following severe respiratory infections.


Assuntos
Proteína ADAMTS4/metabolismo , Fibroblastos/enzimologia , Fibroblastos/patologia , Vírus da Influenza A/patogenicidade , Pulmão/patologia , Pulmão/fisiopatologia , Proteína ADAMTS4/antagonistas & inibidores , Animais , Aves/virologia , Matriz Extracelular/enzimologia , Perfilação da Expressão Gênica , Humanos , Influenza Aviária/virologia , Influenza Humana/patologia , Influenza Humana/terapia , Influenza Humana/virologia , Interferons/imunologia , Interferons/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Pulmão/enzimologia , Pulmão/virologia , Camundongos , Síndrome do Desconforto Respiratório/enzimologia , Síndrome do Desconforto Respiratório/fisiopatologia , Síndrome do Desconforto Respiratório/terapia , Síndrome do Desconforto Respiratório/virologia , Estações do Ano , Análise de Célula Única , Células Estromais/metabolismo
9.
EMBO J ; 40(6): e105543, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33586810

RESUMO

Influenza A virus (IAV) and SARS-CoV-2 (COVID-19) cause pandemic infections where cytokine storm syndrome and lung inflammation lead to high mortality. Given the high social and economic cost of respiratory viruses, there is an urgent need to understand how the airways defend against virus infection. Here we use mice lacking the WD and linker domains of ATG16L1 to demonstrate that ATG16L1-dependent targeting of LC3 to single-membrane, non-autophagosome compartments - referred to as non-canonical autophagy - protects mice from lethal IAV infection. Mice with systemic loss of non-canonical autophagy are exquisitely sensitive to low-pathogenicity IAV where extensive viral replication throughout the lungs, coupled with cytokine amplification mediated by plasmacytoid dendritic cells, leads to fulminant pneumonia, lung inflammation and high mortality. IAV was controlled within epithelial barriers where non-canonical autophagy reduced IAV fusion with endosomes and activation of interferon signalling. Conditional mouse models and ex vivo analysis showed that protection against IAV infection of lung was independent of phagocytes and other leucocytes. This establishes non-canonical autophagy in airway epithelial cells as a novel innate defence that restricts IAV infection and lethal inflammation at respiratory surfaces.


Assuntos
Proteínas Relacionadas à Autofagia/genética , Vírus da Influenza A/patogenicidade , Proteínas Associadas aos Microtúbulos/metabolismo , Infecções por Orthomyxoviridae/genética , Deleção de Sequência , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/virologia , Animais , Autofagia , Proteínas Relacionadas à Autofagia/química , Proteínas Relacionadas à Autofagia/metabolismo , Embrião de Galinha , Citocinas/metabolismo , Cães , Células Madin Darby de Rim Canino , Camundongos , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/mortalidade , Domínios Proteicos , Replicação Viral
10.
Immunity ; 44(1): 46-58, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26789921

RESUMO

Viruses are obligate parasites and thus require the machinery of the host cell to replicate. Inhibition of host factors co-opted during active infection is a strategy hosts use to suppress viral replication and a potential pan-antiviral therapy. To define the cellular proteins and processes required for a virus during infection is thus crucial to understanding the mechanisms of virally induced disease. In this report, we generated fully infectious tagged influenza viruses and used infection-based proteomics to identify pivotal arms of cellular signaling required for influenza virus growth and infectivity. Using mathematical modeling and genetic and pharmacologic approaches, we revealed that modulation of Sec61-mediated cotranslational translocation selectively impaired glycoprotein proteostasis of influenza as well as HIV and dengue viruses and led to inhibition of viral growth and infectivity. Thus, by studying virus-human protein-protein interactions in the context of active replication, we have identified targetable host factors for broad-spectrum antiviral therapies.


Assuntos
Interações Hospedeiro-Parasita/fisiologia , Vírus da Influenza A/fisiologia , Vírus da Influenza A/patogenicidade , Modelos Teóricos , Replicação Viral/fisiologia , Vírus da Dengue/patogenicidade , Vírus da Dengue/fisiologia , HIV/patogenicidade , HIV/fisiologia , Humanos , Imunoprecipitação , Espectrometria de Massas , Dobramento de Proteína , Proteômica
11.
Rev Med Virol ; 34(4): e2559, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38886173

RESUMO

The World Organization for Animal Health defines Avian Influenza Virus as a highly infectious disease caused by diverse subtypes that continue to evolve rapidly, impacting poultry species, pet birds, wild birds, non-human mammals, and occasionally humans. The effects of Avian influenza viruses have been recognised as a precursor for serious health concerns among affected birds, poultry, and human populations in the Middle East. Furthermore, low and high pathogenic avian influenza viruses lead to respiratory illness with varying severity, depending on the virus subtype (e.g., H5, H7, H9, etc.). Possible future outbreaks and endemics of newly emerging subtypes are expected to occur, as many studies have reported the emergence of novel mutations and viral subtypes. However, proper surveillance programs and biosecurity applications should be developed, and countries with incapacitated defences against such outbreaks should be encouraged to undergo complete reinstation and reinforcement in their health and research sectors. Public education regarding biosafety and virus prevention is necessary to ensure minimal spread of avian influenza endemic.


Assuntos
Aves , Vírus da Influenza A , Influenza Aviária , Influenza Humana , Animais , Influenza Aviária/epidemiologia , Influenza Aviária/virologia , Influenza Aviária/prevenção & controle , Influenza Aviária/transmissão , Humanos , Influenza Humana/prevenção & controle , Influenza Humana/epidemiologia , Influenza Humana/virologia , Região do Mediterrâneo/epidemiologia , Aves/virologia , Vírus da Influenza A/genética , Vírus da Influenza A/fisiologia , Vírus da Influenza A/patogenicidade , Surtos de Doenças/prevenção & controle , Surtos de Doenças/veterinária
12.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-34969849

RESUMO

Infection by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) provokes a potentially fatal pneumonia with multiorgan failure, and high systemic inflammation. To gain mechanistic insight and ferret out the root of this immune dysregulation, we modeled, by in vitro coculture, the interactions between infected epithelial cells and immunocytes. A strong response was induced in monocytes and B cells, with a SARS-CoV-2-specific inflammatory gene cluster distinct from that seen in influenza A or Ebola virus-infected cocultures, and which reproduced deviations reported in blood or lung myeloid cells from COVID-19 patients. A substantial fraction of the effect could be reproduced after individual transfection of several SARS-CoV-2 proteins (Spike and some nonstructural proteins), mediated by soluble factors, but not via transcriptional induction. This response was greatly muted in monocytes from healthy children, perhaps a clue to the age dependency of COVID-19. These results suggest that the inflammatory malfunction in COVID-19 is rooted in the earliest perturbations that SARS-CoV-2 induces in epithelia.


Assuntos
COVID-19/imunologia , Células Epiteliais/imunologia , Monócitos/imunologia , SARS-CoV-2/patogenicidade , Adulto , Linfócitos B/imunologia , COVID-19/patologia , Criança , Técnicas de Cocultura , Ebolavirus/patogenicidade , Células Epiteliais/virologia , Perfilação da Expressão Gênica , Humanos , Inflamação , Vírus da Influenza A/patogenicidade , Pulmão/imunologia , Células Mieloides/imunologia , Especificidade da Espécie , Proteínas Virais/imunologia
13.
J Biol Chem ; 299(1): 102747, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36436557

RESUMO

Myxovirus resistance protein 1 (MX1) and MX2 are homologous, dynamin-like large GTPases, induced upon interferon exposure. Human MX1 (HsMX1) is known to inhibit many viruses, including influenza A virus, by likely acting at various steps of their life cycles. Despite decades of studies, the mechanism(s) of action with which MX1 proteins manage to inhibit target viruses is not fully understood. MX1 proteins are mechano-enzymes and share a similar organization to dynamin, with a GTPase domain and a carboxy-terminal stalk domain, connected by a bundle signaling element. These three elements are known to be essential for antiviral activity. HsMX1 has two unstructured regions, the L4 loop, also essential for antiviral activity, and a short amino (N)-terminal region, which greatly varies between MX1 proteins of different species. The role of this N-terminal domain in antiviral activity is not known. Herein, using mutagenesis, imaging, and biochemical approaches, we demonstrate that the N-terminal domain of HsMX1 is essential for antiviral activity against influenza A virus, Vesicular Stomatitis Virus, and La Crosse virus. Furthermore, we pinpoint a highly conserved leucine within this region, which is absolutely crucial for human, mouse, and bat MX1 protein antiviral activity. Importantly, mutation of this leucine does not compromise GTPase activity or oligomerization capabilities but does modify MX1 protein subcellular localization. The discovery of this essential and highly conserved residue defines this region as key for antiviral activity and may reveal insights as to the mechanism(s) of action of MX1 proteins.


Assuntos
Vírus da Influenza A , Proteínas de Resistência a Myxovirus , Vírus de RNA , Animais , Humanos , Camundongos , Antivirais/farmacologia , Antivirais/metabolismo , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/metabolismo , Vírus da Influenza A/metabolismo , Vírus da Influenza A/patogenicidade , Leucina , Proteínas de Resistência a Myxovirus/genética , Proteínas de Resistência a Myxovirus/metabolismo , Proteínas/metabolismo , Vírus de RNA/metabolismo , Vírus de RNA/patogenicidade
14.
Infect Immun ; 92(5): e0045323, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38602405

RESUMO

Nontypeable Haemophilus influenzae (NTHi) is a major otitis media (OM) pathogen, with colonization a prerequisite for disease development. Most acute OM is in children <5 years old, with recurrent and chronic OM impacting hearing and learning. Therapies to prevent NTHi colonization and/or disease are needed, especially for young children. Respiratory viruses are implicated in driving the development of bacterial OM in children. We have developed an infant mouse model of influenza-driven NTHi OM, as a preclinical tool for the evaluation of safety and efficacy of clinical therapies to prevent NTHi colonization and the development of OM. In this model, 100% of infant BALB/cARC mice were colonized with NTHi, and all developed NTHi OM. Influenza A virus (IAV) facilitated the establishment of dense (1 × 105 CFU/mL) and long-lasting (6 days) NTHi colonization. IAV was essential for the development of NTHi OM, with 100% of mice in the IAV/NTHi group developing NTHi OM compared with 8% of mice in the NTHi only group. Histological analysis and cytokine measurements revealed that the inflammation observed in the middle ear of the infant mice with OM reflected inflammation observed in children with OM. We have developed the first infant mouse model of NTHi colonization and OM. This ascension model uses influenza-driven establishment of OM and reflects the clinical pathology of bacterial OM developing after a respiratory virus infection. This model provides a valuable tool for testing therapies to prevent or treat NTHi colonization and disease in young children.


Assuntos
Modelos Animais de Doenças , Infecções por Haemophilus , Haemophilus influenzae , Vírus da Influenza A , Otite Média , Animais , Otite Média/microbiologia , Haemophilus influenzae/crescimento & desenvolvimento , Haemophilus influenzae/patogenicidade , Haemophilus influenzae/fisiologia , Infecções por Haemophilus/microbiologia , Camundongos , Vírus da Influenza A/patogenicidade , Vírus da Influenza A/crescimento & desenvolvimento , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/virologia , Infecções por Orthomyxoviridae/complicações , Humanos , Animais Recém-Nascidos
15.
J Gen Virol ; 105(7)2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38975739

RESUMO

The 2020/2021 epidemic in Europe of highly pathogenic avian influenza virus (HPAIV) of subtype H5 surpassed all previously recorded European outbreaks in size, genotype constellations and reassortment frequency and continued into 2022 and 2023. The causative 2.3.4.4b viral lineage proved to be highly proficient with respect to reassortment with cocirculating low pathogenic avian influenza viruses and seems to establish an endemic status in northern Europe. A specific HPAIV reassortant of the subtype H5N3 was detected almost exclusively in red knots (Calidris canutus islandica) in December 2020. It caused systemic and rapidly fatal disease leading to a singular and self-limiting mass mortality affecting about 3500 birds in the German Wadden Sea, roughly 1 % of the entire flyway population of islandica red knots. Phylogenetic analyses revealed that the H5N3 reassortant very likely had formed in red knots and remained confined to this species. While mechanisms of virus circulation in potential reservoir species, dynamics of spill-over and reassortment events and the roles of environmental virus sources remain to be identified, the year-round infection pressure poses severe threats to endangered avian species and prompts adaptation of habitat and species conservation practices.


Assuntos
Vírus da Influenza A , Influenza Aviária , Filogenia , Vírus Reordenados , Animais , Influenza Aviária/virologia , Influenza Aviária/epidemiologia , Europa (Continente)/epidemiologia , Vírus da Influenza A/genética , Vírus da Influenza A/classificação , Vírus da Influenza A/patogenicidade , Vírus Reordenados/genética , Surtos de Doenças/veterinária , Charadriiformes/virologia , Aves/virologia
16.
Am J Physiol Heart Circ Physiol ; 327(4): H937-H946, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39150394

RESUMO

Influenza A virus (IAV) infection while primarily affecting the lungs, is often associated with cardiovascular complications. However, the mechanisms underlying this association are not fully understood. Here, we investigated the potential role of FBXL19, a member of the Skp1-Cullin-1-F-box family of E3 ubiquitin ligase, in IAV-induced cardiac inflammation. We demonstrated that FBXL19 overexpression in endothelial cells (ECs) reduced viral titers and IAV matrix protein 1 (M1) levels while increasing antiviral gene expression, including interferon (IFN)-α, -ß, and -γ and RANTES (regulated on activation normal T cell expressed and secreted) in the cardiac tissue of IAV-infected mice. Moreover, EC-specific overexpression of FBXL19 attenuated the IAV infection-reduced interferon regulatory factor 3 (IRF3) level without altering its mRNA level and suppressed cardiac inflammation. Furthermore, IAV infection triggered cellular senescence programs in the heart as indicated by the upregulation of p16 and p21 mRNA levels and the downregulation of lamin-B1 levels, which were partially reversed by FBXL19 overexpression in ECs. Our findings indicate that EC-specific overexpression of FBXL19 protects against IAV-induced cardiac damage by enhancing interferon-mediated antiviral signaling, reducing cardiac inflammation, and suppressing cellular senescence programs.NEW & NOTEWORTHY Our study reveals a novel facet of IAV infection, demonstrating that it can trigger cellular senescence within the heart. Intriguingly, upregulation of endothelial FBXL19 promotes host innate immunity, reduces cardiac senescence, and diminishes inflammation. These findings highlight the therapeutic potential of targeting FBXL19 to mitigate IAV-induced cardiovascular complications.


Assuntos
Senescência Celular , Células Endoteliais , Fator Regulador 3 de Interferon , Infecções por Orthomyxoviridae , Animais , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/imunologia , Células Endoteliais/virologia , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/genética , Camundongos Endogâmicos C57BL , Camundongos , Proteínas F-Box/metabolismo , Proteínas F-Box/genética , Humanos , Vírus da Influenza A/patogenicidade , Miocárdio/metabolismo , Miocárdio/imunologia , Miocárdio/patologia , Modelos Animais de Doenças , Transdução de Sinais , Interferons/metabolismo , Interferons/genética , Masculino , Quimiocina CCL5
17.
J Virol ; 97(1): e0153622, 2023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36602361

RESUMO

As influenza A viruses (IAV) continue to cross species barriers and cause human infection, the establishment of risk assessment rubrics has improved pandemic preparedness efforts. In vivo pathogenicity and transmissibility evaluations in the ferret model represent a critical component of this work. As the relative contribution of in vitro experimentation to these rubrics has not been closely examined, we sought to evaluate to what extent viral titer measurements over the course of in vitro infections are predictive or correlates of nasal wash and tissue measurements for IAV infections in vivo. We compiled data from ferrets inoculated with an extensive panel of over 50 human and zoonotic IAV (inclusive of swine-origin and high- and low-pathogenicity avian influenza viruses associated with human infection) under a consistent protocol, with all viruses concurrently tested in a human bronchial epithelial cell line (Calu-3). Viral titers in ferret nasal wash specimens and nasal turbinate tissue correlated positively with peak titer in Calu-3 cells, whereas additional phenotypic and molecular determinants of influenza virus virulence and transmissibility in ferrets varied in their association with in vitro viral titer measurements. Mathematical modeling was used to estimate more generalizable key replication kinetic parameters from raw in vitro viral titers, revealing commonalities between viral infection progression in vivo and in vitro. Meta-analyses inclusive of IAV that display a diverse range of phenotypes in ferrets, interpreted with mathematical modeling of viral kinetic parameters, can provide critical information supporting a more rigorous and appropriate contextualization of in vitro experiments toward pandemic preparedness. IMPORTANCE Both in vitro and in vivo models are employed for assessing the pandemic potential of novel and emerging influenza A viruses in laboratory settings, but systematic examinations of how well viral titer measurements obtained in vitro align with results from in vivo experimentation are not frequently performed. We show that certain viral titer measurements following infection of a human bronchial epithelial cell line are positively correlated with viral titers in specimens collected from virus-inoculated ferrets and employ mathematical modeling to identify commonalities between viral infection progression between both models. These analyses provide a necessary first step in enhanced interpretation and incorporation of in vitro-derived data in risk assessment activities and highlight the utility of employing mathematical modeling approaches to more closely examine features of virus replication not identifiable by experimental studies alone.


Assuntos
Vírus da Influenza A , Infecções por Orthomyxoviridae , Medição de Risco , Animais , Humanos , Furões , Vírus da Influenza A/patogenicidade , Influenza Humana , Infecções por Orthomyxoviridae/patologia , Medição de Risco/métodos , Suínos , Replicação Viral , Linhagem Celular , Técnicas In Vitro
18.
PLoS Pathog ; 18(4): e1010446, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35377920

RESUMO

Host defense systems employ posttranslational modifications to protect against invading pathogens. Here, we found that protein inhibitor of activated STAT 1 (PIAS1) interacts with the nucleoprotein (NP), polymerase basic protein 1 (PB1), and polymerase basic protein 2 (PB2) of influenza A virus (IAV). Lentiviral-mediated stable overexpression of PIAS1 dramatically suppressed the replication of IAV, whereas siRNA knockdown or CRISPR/Cas9 knockout of PIAS1 expression significantly increased virus growth. The expression of PIAS1 was significantly induced upon IAV infection in both cell culture and mice, and PIAS1 was involved in the overall increase in cellular SUMOylation induced by IAV infection. We found that PIAS1 inhibited the activity of the viral RNP complex, whereas the C351S or W372A mutant of PIAS1, which lacks the SUMO E3 ligase activity, lost the ability to suppress the activity of the viral RNP complex. Notably, the SUMO E3 ligase activity of PIAS1 catalyzed robust SUMOylation of PB2, but had no role in PB1 SUMOylation and a minimal role in NP SUMOylation. Moreover, PIAS1-mediated SUMOylation remarkably reduced the stability of IAV PB2. When tested in vivo, we found that the downregulation of Pias1 expression in mice enhanced the growth and virulence of IAV. Together, our findings define PIAS1 as a restriction factor for the replication and pathogenesis of IAV.


Assuntos
Vírus da Influenza A , Proteínas Inibidoras de STAT Ativados , Sumoilação , Replicação Viral , Animais , Vírus da Influenza A/patogenicidade , Vírus da Influenza A/fisiologia , Camundongos , Proteínas Inibidoras de STAT Ativados/genética , Proteínas Inibidoras de STAT Ativados/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Virulência
19.
Acta Neuropathol ; 147(1): 77, 2024 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-38687393

RESUMO

Influenza-associated encephalopathy (IAE) is extremely acute in onset, with high lethality and morbidity within a few days, while the direct pathogenesis by influenza virus in this acute phase in the brain is largely unknown. Here we show that influenza virus enters into the cerebral endothelium and thereby induces IAE. Three-weeks-old young mice were inoculated with influenza A virus (IAV). Physical and neurological scores were recorded and temporal-spatial analyses of histopathology and viral studies were performed up to 72 h post inoculation. Histopathological examinations were also performed using IAE human autopsy brains. Viral infection, proliferation and pathogenesis were analyzed in cell lines of endothelium and astrocyte. The effects of anti-influenza viral drugs were tested in the cell lines and animal models. Upon intravenous inoculation of IAV in mice, the mice developed encephalopathy with brain edema and pathological lesions represented by micro bleeding and injured astrocytic process (clasmatodendrosis) within 72 h. Histologically, massive deposits of viral nucleoprotein were observed as early as 24 h post infection in the brain endothelial cells of mouse models and the IAE patients. IAV inoculated endothelial cell lines showed deposition of viral proteins and provoked cell death, while IAV scarcely amplified. Inhibition of viral transcription and translation suppressed the endothelial cell death and the lethality of mouse models. These data suggest that the onset of encephalopathy should be induced by cerebral endothelial infection with IAV. Thus, IAV entry into the endothelium, and transcription and/or translation of viral RNA, but not viral proliferation, should be the key pathogenesis of IAE.


Assuntos
Encéfalo , Infecções por Orthomyxoviridae , Animais , Humanos , Camundongos , Encéfalo/patologia , Encéfalo/virologia , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Infecções por Orthomyxoviridae/complicações , Internalização do Vírus , Vírus da Influenza A/patogenicidade , Células Endoteliais/virologia , Células Endoteliais/patologia , Influenza Humana/patologia , Influenza Humana/complicações , Encefalopatias/virologia , Encefalopatias/patologia , Masculino , Modelos Animais de Doenças , Feminino , Endotélio/patologia , Endotélio/virologia , Camundongos Endogâmicos C57BL
20.
Avian Pathol ; 53(4): 242-246, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38345041

RESUMO

Based on the pathogenicity in chickens, most H1-H16 avian influenza viruses (AIV) cause mild diseases, whereas some of the H5 and H7 AI viruses cause severe, systemic disease. The number of basic amino acids in the haemagglutinin (HA) cleavage site of AIV plays a critical role in pathogenicity. As we gain a greater understanding of the molecular mechanisms of pathogenicity, genome sequencing of the HA0 cleavage site has assumed a greater role in assessment of the potential pathogenicity of H5 and H7 viruses. We validated the use of HA cleavage site motif analysis by comparing molecular pathotyping data against experimental in vivo (intravenous pathogenicity index [IVPI] and lethality) data for determination of both low pathogenicity and high pathogenicity AI virus declaration with the goal of expediting pathotype confirmation and further reducing the reliance on in vivo testing. Our data provide statistical support to the continued use of molecular determination of pathotype for AI viruses based on the HA cleavage site sequence in the absence of an in vivo study determination. This approach not only expedites the declaration process of highly pathogenic AIV (HPAIV) but also reduces the need for experimental in vivo testing of H5 and H7 viruses.


Assuntos
Galinhas , Genoma Viral , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Vírus da Influenza A , Influenza Aviária , Animais , Influenza Aviária/virologia , Galinhas/virologia , Vírus da Influenza A/patogenicidade , Vírus da Influenza A/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Virulência , Fenótipo , Doenças das Aves Domésticas/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA