Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 210(3): 322-334, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36525001

RESUMO

Human macrophages secrete extracellular vesicles (EVs) loaded with numerous immunoregulatory proteins. Vesicle-mediated protein secretion in macrophages is regulated by poorly characterized mechanisms; however, it is now known that inflammatory conditions significantly alter both the quantities and protein composition of secreted vesicles. In this study, we employed high-throughput quantitative proteomics to characterize the modulation of EV-mediated protein secretion during noncanonical caspase-4/5 inflammasome activation via LPS transfection. We show that human macrophages activate robust caspase-4-dependent EV secretion upon transfection of LPS, and this process is also partially dependent on NLRP3 and caspase-5. A similar effect occurs with delivery of the LPS with Escherichia coli-derived outer membrane vesicles. Moreover, sensitization of the macrophages through TLR4 by LPS priming prior to LPS transfection dramatically augments the EV-mediated protein secretion. Our data demonstrate that this process differs significantly from canonical inflammasome activator ATP-induced vesiculation, and it is dependent on the autocrine IFN signal associated with TLR4 activation. LPS priming preceding the noncanonical inflammasome activation significantly enhances vesicle-mediated secretion of inflammasome components caspase-1, ASC, and lytic cell death effectors GSDMD, MLKL, and NINJ1, suggesting that inflammatory EV transfer may exert paracrine effects in recipient cells. Moreover, using bioinformatics methods, we identify 15-deoxy-Δ12,14-PGJ2 and parthenolide as inhibitors of caspase-4-mediated inflammation and vesicle secretion, indicating new therapeutic potential of these anti-inflammatory drugs.


Assuntos
Vesículas Extracelulares , Lipopolissacarídeos , Macrófagos , Humanos , Caspases/metabolismo , Escherichia coli/metabolismo , Vesículas Extracelulares/metabolismo , Inflamassomos/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , Fatores de Crescimento Neural/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Receptor 4 Toll-Like/metabolismo
2.
J Immunol ; 204(12): 3063-3069, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32513874

RESUMO

Inflammasomes are multiprotein complexes of the innate immune system that orchestrate development of inflammation by activating the secretion of proinflammatory cytokines, IL-1ß and IL-18. The LPS of Gram-negative bacteria have been shown to activate a novel, noncanonical inflammasome by directly binding in the cytosol to human caspase-4 and mouse caspase-11. Activation of noncanonical inflammasome exerts two major effects: it activates the NLRP3-caspase-1-mediated processing and secretion of IL-1ß and IL-18 and induces the inflammatory cell death, pyroptosis, via gasdermin D. This previously unexpected cytosolic LPS sensing of the innate immune system provides critical hints for host response to Gram-negative bacterial infections and development of different inflammatory diseases. However, many of its molecular regulatory mechanisms are yet to be discovered. In this review, we provide comprehensive analysis of current understanding of intracellular LPS detection and pyroptosis via noncanonical inflammasome and discuss the recently proposed mechanisms of its function and regulation.


Assuntos
Caspases/metabolismo , Inflamassomos/metabolismo , Lipopolissacarídeos/metabolismo , Animais , Humanos , Inflamação/metabolismo , Piroptose/fisiologia
3.
EMBO J ; 35(13): 1385-99, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27234299

RESUMO

Herpes simplex virus (HSV) 1 stimulates type I IFN expression through the cGAS-STING-TBK1 signaling axis. Macrophages have recently been proposed to be an essential source of IFN during viral infection. However, it is not known how HSV-1 inhibits IFN expression in this cell type. Here, we show that HSV-1 inhibits type I IFN induction through the cGAS-STING-TBK1 pathway in human macrophages, in a manner dependent on the conserved herpesvirus protein ICP27. This viral protein was expressed de novo in macrophages with early nuclear localization followed by later translocation to the cytoplasm where ICP27 prevented activation of IRF3. ICP27 interacted with TBK1 and STING in a manner that was dependent on TBK1 activity and the RGG motif in ICP27. Thus, HSV-1 inhibits expression of type I IFN in human macrophages through ICP27-dependent targeting of the TBK1-activated STING signalsome.


Assuntos
Herpesvirus Humano 1/patogenicidade , Proteínas Imediatamente Precoces/metabolismo , Evasão da Resposta Imune , Interferon Tipo I/antagonistas & inibidores , Macrófagos/imunologia , Proteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Células Cultivadas , Interações Hospedeiro-Patógeno , Humanos , Mapeamento de Interação de Proteínas
4.
Mol Cell Proteomics ; 16(4 suppl 1): S187-S199, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28196878

RESUMO

Gram-negative bacteria are associated with a wide spectrum of infectious diseases in humans. Inflammasomes are cytosolic protein complexes that are assembled when the cell encounters pathogens or other harmful agents. The non-canonical caspase-4/5 inflammasome is activated by Gram-negative bacteria-derived lipopolysaccharide (LPS) and by endogenous oxidized phospholipids. Protein secretion is a critical component of the innate immune response. Here, we have used label-free quantitative proteomics to characterize global protein secretion in response to non-canonical inflammasome activation upon intracellular LPS recognition in human primary macrophages. Before proteomics, the total secretome was separated into two fractions, enriched extracellular vesicle (EV) fraction and rest-secretome (RS) fraction using size-exclusion centrifugation. We identified 1048 proteins from the EV fraction and 1223 proteins from the RS fraction. From these, 640 were identified from both fractions suggesting that the non-canonical inflammasome activates multiple, partly overlapping protein secretion pathways. We identified several secreted proteins that have a critical role in host response against severe Gram-negative bacterial infection. The soluble secretome (RS fraction) was highly enriched with inflammation-associated proteins upon intracellular LPS recognition. Several ribosomal proteins were highly abundant in the EV fraction upon infection, and our data strongly suggest that secretion of translational machinery and concomitant inhibition of translation are important parts of host response against Gram-negative bacteria sensing caspase-4/5 inflammasome. Intracellular recognition of LPS resulted in the secretion of two metalloproteinases, adisintegrin and metalloproteinase domain-containing protein 10 (ADAM10) and MMP14, in the enriched EV fraction. ADAM10 release was associated with the secretion of TNF, a key inflammatory cytokine, and M-CSF, an important growth factor for myeloid cells probably through ADAM10-dependent membrane shedding of these cytokines. Caspase-4/5 inflammasome activation also resulted in secretion of danger-associated molecules S100A8 and prothymosin-α in the enriched EV fraction. Both S100A8 and prothymosin-α are ligands for toll-like receptor 4 recognizing extracellular LPS, and they may contribute to endotoxic shock during non-canonical inflammasome activation.


Assuntos
Inflamassomos/metabolismo , Lipopolissacarídeos/efeitos adversos , Macrófagos/metabolismo , Proteoma/metabolismo , Proteína ADAM10/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Caspases/metabolismo , Caspases Iniciadoras/metabolismo , Células Cultivadas , Infecções por Bactérias Gram-Negativas/imunologia , Humanos , Imunidade Inata , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Metaloproteinase 14 da Matriz/metabolismo , Proteínas de Membrana/metabolismo , Proteômica/métodos
5.
FASEB J ; 31(11): 4770-4782, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28705811

RESUMO

Invading pathogens provoke robust innate immune responses in Dipteran insects, such as Drosophila melanogaster In a systemic bacterial infection, a humoral response is induced in the fat body. Gram-positive bacteria trigger the Toll signaling pathway, whereas gram-negative bacterial infections are signaled via the immune deficiency (IMD) pathway. We show here that the RNA interference-mediated silencing of Furin1-a member of the proprotein convertase enzyme family-specifically in the fat body, results in a reduction in the expression of antimicrobial peptides. This, in turn, compromises the survival of adult fruit flies in systemic infections that are caused by both gram-positive and -negative bacteria. Furin1 plays a nonredundant role in the regulation of immune responses, as silencing of Furin2, the other member of the enzyme family, had no effect on survival or the expression of antimicrobial peptides upon a systemic infection. Furin1 does not directly affect the Toll or IMD signaling pathways, but the reduced expression of Furin1 up-regulates stress response factors in the fat body. We also demonstrate that Furin1 is a negative regulator of the Janus kinase/signal transducer and activator of transcription signaling pathway, which is implicated in stress responses in the fly. In summary, our data identify Furin1 as a novel regulator of humoral immunity and cellular stress responses in Drosophila-Aittomäki, S., Valanne, S., Lehtinen, T., Matikainen, S., Nyman, T. A., Rämet, M., Pesu, M. Proprotein convertase Furin1 expression in the Drosophila fat body is essential for a normal antimicrobial peptide response and bacterial host defense.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteínas de Drosophila/biossíntese , Furina/biossíntese , Regulação Enzimológica da Expressão Gênica , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/imunologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/imunologia , Drosophila melanogaster , Furina/genética , Furina/imunologia , Bactérias Gram-Negativas/imunologia , Bactérias Gram-Negativas/metabolismo , Infecções por Bactérias Gram-Negativas/enzimologia , Infecções por Bactérias Gram-Negativas/genética , Infecções por Bactérias Gram-Negativas/imunologia , Bactérias Gram-Positivas/imunologia , Bactérias Gram-Positivas/metabolismo , Infecções por Bactérias Gram-Positivas/enzimologia , Infecções por Bactérias Gram-Positivas/genética , Infecções por Bactérias Gram-Positivas/imunologia
6.
J Immunol ; 197(8): 3315-3325, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27638862

RESUMO

Extracellular ATP is an endogenous danger signal that is known to activate inflammatory responses in innate immune cells, including macrophages. Activated macrophages start to secrete proteins to induce an immune response, as well as to recruit other immune cells to the site of infection and tissue damage. In this study, we characterized the secretome (i.e., the global pattern of secreted proteins) of ATP-stimulated human macrophages. We show that ATP stimulation activates robust vesicle-mediated unconventional protein secretion, including exosome release and membrane shedding, from human macrophages. Pathway analysis of the identified secreted proteins showed that calpain-related pathways were overrepresented in the secretome of ATP-stimulated cells. In accordance with this, calpains, which are calcium-dependent nonlysosomal cysteine proteases, were activated upon ATP stimulation through a P2X purinoceptor 7 receptor-dependent pathway. Functional studies demonstrated that calpain activity is essential for the P2X purinoceptor 7 receptor-mediated activation of unconventional protein secretion. Unconventional protein secretion was followed by cell necrosis and NLRP3 inflammasome-mediated secretion of the mature form of the proinflammatory cytokine IL-1ß. Furthermore, ATP-driven NLRP3 inflammasome activation was also dependent on calpain activity. Interestingly, pro-IL-1ß and inflammasome components ASC and caspase-1 were released by ATP-activated macrophages through a vesicle-mediated secretion pathway. In conclusion, to our knowledge, we provide the first global characterization of proteins secreted by ATP-activated human macrophages and show a pivotal role for calpains in the activation of the inflammatory response during ATP exposure.


Assuntos
Trifosfato de Adenosina/metabolismo , Calpaína/metabolismo , Inflamassomos/metabolismo , Macrófagos/metabolismo , Humanos , Inflamassomos/imunologia , Macrófagos/imunologia
7.
Mol Cell Proteomics ; 15(10): 3203-3219, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27486199

RESUMO

Influenza A viruses cause infections in the human respiratory tract and give rise to annual seasonal outbreaks, as well as more rarely dreaded pandemics. Influenza A viruses become quickly resistant to the virus-directed antiviral treatments, which are the current main treatment options. A promising alternative approach is to target host cell factors that are exploited by influenza viruses. To this end, we characterized the phosphoproteome of influenza A virus infected primary human macrophages to elucidate the intracellular signaling pathways and critical host factors activated upon influenza infection. We identified 1675 phosphoproteins, 4004 phosphopeptides and 4146 nonredundant phosphosites. The phosphorylation of 1113 proteins (66%) was regulated upon infection, highlighting the importance of such global phosphoproteomic profiling in primary cells. Notably, 285 of the identified phosphorylation sites have not been previously described in publicly available phosphorylation databases, despite many published large-scale phosphoproteome studies using human and mouse cell lines. Systematic bioinformatics analysis of the phosphoproteome data indicated that the phosphorylation of proteins involved in the ubiquitin/proteasome pathway (such as TRIM22 and TRIM25) and antiviral responses (such as MAVS) changed in infected macrophages. Proteins known to play roles in small GTPase-, mitogen-activated protein kinase-, and cyclin-dependent kinase- signaling were also regulated by phosphorylation upon infection. In particular, the influenza infection had a major influence on the phosphorylation profiles of a large number of cyclin-dependent kinase substrates. Functional studies using cyclin-dependent kinase inhibitors showed that the cyclin-dependent kinase activity is required for efficient viral replication and for activation of the host antiviral responses. In addition, we show that cyclin-dependent kinase inhibitors protect IAV-infected mice from death. In conclusion, we provide the first comprehensive phosphoproteome characterization of influenza A virus infection in primary human macrophages, and provide evidence that cyclin-dependent kinases represent potential therapeutic targets for more effective treatment of influenza infections.


Assuntos
Vírus da Influenza A/patogenicidade , Influenza Humana/metabolismo , Macrófagos/virologia , Fosfoproteínas/análise , Proteômica/métodos , Animais , Biologia Computacional/métodos , Quinases Ciclina-Dependentes/metabolismo , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Macrófagos/metabolismo , Camundongos , Transdução de Sinais
8.
J Proteome Res ; 16(1): 217-227, 2017 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-27723984

RESUMO

Influenza A viruses (IAVs) are aggressive pathogens that cause acute respiratory diseases and annual epidemics in humans. Host defense against IAV infection is initiated by macrophages, which are the principal effector cells of the innate immune system. We have previously shown that IAV infection of human macrophages is associated with robust secretion of proteins via conventional and unconventional protein release pathways. Here we have characterized unconventional, extracellular vesicle (EV)-mediated protein secretion in human macrophages during IAV infection using proteomics, bioinformatics, and functional studies. We demonstrate that at 9 h postinfection a robust EV-mediated protein secretion takes place. We identified 2359 human proteins from EVs of IAV-infected macrophages compared with 1448 proteins identified from EVs of control cells. Bioinformatic analysis shows that many proteins involved in translation, like components of spliceosome machinery and the ribosome, are secreted in EVs in response to IAV infection. Our data also shows that EVs derived from IAV-infected macrophages contain fatty acid-binding proteins, antiviral cytokines, copper metabolism Murr-1 domain proteins, and autophagy-related proteins. In addition, our data suggest that secretory autophagy plays a role in activating EV-mediated protein secretion during IAV infection.


Assuntos
Vesículas Extracelulares/genética , Interações Hospedeiro-Patógeno , Vírus da Influenza A Subtipo H3N2/fisiologia , Macrófagos/metabolismo , Proteoma/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Relacionadas à Autofagia/genética , Proteínas Relacionadas à Autofagia/imunologia , Proteínas Relacionadas à Autofagia/metabolismo , Biologia Computacional , Citocinas/genética , Citocinas/imunologia , Citocinas/metabolismo , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/virologia , Proteínas de Ligação a Ácido Graxo/genética , Proteínas de Ligação a Ácido Graxo/imunologia , Proteínas de Ligação a Ácido Graxo/metabolismo , Regulação da Expressão Gênica , Ontologia Genética , Humanos , Macrófagos/imunologia , Macrófagos/virologia , Anotação de Sequência Molecular , Cultura Primária de Células , Biossíntese de Proteínas , Proteoma/imunologia , Proteoma/metabolismo , Transdução de Sinais
9.
Biochim Biophys Acta ; 1859(11): 1440-1448, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27664935

RESUMO

Influenza NS1 protein is an important virulence factor that is capable of binding double-stranded (ds) RNA and inhibiting dsRNA-mediated host innate immune responses. Here we show that NS1 can also bind cellular dsDNA. This interaction prevents loading of transcriptional machinery to the DNA, thereby attenuating IAV-mediated expression of antiviral genes. Thus, we identified a previously undescribed strategy, by which RNA virus inhibits cellular transcription to escape antiviral response and secure its replication.


Assuntos
DNA/metabolismo , Transcrição Gênica/fisiologia , Proteínas não Estruturais Virais/metabolismo , Animais , Linhagem Celular , Cromatina/metabolismo , Humanos , Vírus da Influenza A/fisiologia , Ligação Proteica , Proteínas não Estruturais Virais/fisiologia , Replicação Viral
10.
Expert Rev Proteomics ; 14(5): 395-407, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28406322

RESUMO

INTRODUCTION: The immune system is our defense system against microbial infections and tissue injury, and understanding how it works in detail is essential for developing drugs for different diseases. Mass spectrometry-based proteomics can provide in-depth information on the molecular mechanisms involved in immune responses. Areas covered: Summarized are the key immunology findings obtained with MS-based proteomics in the past five years, with a focus on inflammasome activation, global protein secretion, mucosal immunology, immunopeptidome and T cells. Special focus is on extracellular vesicle-mediated protein secretion and its role in immune responses. Expert commentary: Proteomics is an essential part of modern omics-scale immunology research. To date, MS-based proteomics has been used in immunology to study protein expression levels, their subcellular localization, secretion, post-translational modifications, and interactions in immune cells upon activation by different stimuli. These studies have made major contributions to understanding the molecular mechanisms involved in innate and adaptive immune responses. New developments in proteomics offer constantly novel possibilities for exploring the immune system. Examples of these techniques include mass cytometry and different MS-based imaging approaches which can be widely used in immunology.


Assuntos
Imunidade Inata , Inflamassomos/química , Proteoma/imunologia , Via Secretória , Humanos , Inflamassomos/imunologia , Espectrometria de Massas/métodos , Proteoma/química , Linfócitos T/imunologia
11.
J Immunol ; 192(12): 5952-62, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24808366

RESUMO

Dectin-1 is a membrane-bound pattern recognition receptor for ß-glucans, which are the main constituents of fungal cell walls. Detection of ß-glucans by dectin-1 triggers an effective innate immune response. In this study, we have used a systems biology approach to provide the first comprehensive characterization of the secretome and associated intracellular signaling pathways involved in activation of dectin-1/Syk in human macrophages. Transcriptome and secretome analysis revealed that the dectin-1 pathway induced significant gene expression changes and robust protein secretion in macrophages. The enhanced protein secretion correlated only partly with increased gene expression. Bioinformatics combined with functional studies revealed that the dectin-1/Syk pathway activates both conventional and unconventional, vesicle-mediated, protein secretion. The unconventional protein secretion triggered by the dectin-1 pathway is dependent on inflammasome activity and an active autophagic process. In conclusion, our results reveal that unconventional protein secretion has an important role in the innate immune response against fungal infections.


Assuntos
Autofagia/imunologia , Regulação da Expressão Gênica/imunologia , Imunidade Inata , Inflamassomos/imunologia , Lectinas Tipo C/imunologia , Macrófagos/imunologia , Feminino , Humanos , Inflamassomos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Macrófagos/metabolismo , Masculino , Micoses/imunologia , Micoses/metabolismo , Proteínas Tirosina Quinases/imunologia , Proteínas Tirosina Quinases/metabolismo , Quinase Syk
12.
Mol Cell Proteomics ; 13(10): 2604-17, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24997996

RESUMO

Viral double-stranded RNA (dsRNA) is the most important viral structure recognized by cytosolic pattern-recognition receptors of the innate immune system, and its recognition results in the activation of signaling cascades that stimulate the production of antiviral cytokines and apoptosis of infected cells. 14-3-3 proteins are ubiquitously expressed regulatory molecules that participate in a variety of cellular processes, and 14-3-3 protein-mediated signaling pathways are activated by cytoplasmic dsRNA in human keratinocytes. However, the functional role of 14-3-3 protein-mediated interactions during viral dsRNA stimulation has remained uncharacterized. Here, we used functional proteomics to identify proteins whose phosphorylation and interaction with 14-3-3 is modulated by dsRNA and to characterize the signaling pathways activated during cytosolic dsRNA-induced innate immune response in human HaCaT keratinocytes. Phosphoproteome analysis showed that several MAPK- and immune-response-related signaling pathways were activated after dsRNA stimulation. Interactome analysis identified RelA-associated inhibitor, high-mobility group proteins, and several proteins associated with host responses to viral infection as novel 14-3-3 target proteins. Functional studies showed that RelA-associated inhibitor regulated dsRNA-induced apoptosis and TNF production. Integrated network analyses of proteomic data revealed that sirtuin1 was a central molecule regulated by 14-3-3s during dsRNA stimulation. Further experiments showed that sirtuin 1 negatively regulated dsRNA-induced NFκB transcriptional activity, suppressed expression of antiviral cytokines, and protected cells from apoptosis in dsRNA-stimulated and encephalomyocarditis-virus-infected keratinocytes. In conclusion, our data highlight the importance of 14-3-3 proteins in antiviral responses and identify RelA-associated inhibitor and sirtuin 1 as novel regulators of antiviral innate immune responses.


Assuntos
Proteínas 14-3-3/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Queratinócitos/metabolismo , Proteômica/métodos , RNA de Cadeia Dupla/metabolismo , Proteínas Repressoras/metabolismo , Sirtuína 1/metabolismo , Infecções por Cardiovirus/imunologia , Infecções por Cardiovirus/metabolismo , Linhagem Celular , Citosol/metabolismo , Vírus da Encefalomiocardite/genética , Vírus da Encefalomiocardite/imunologia , Humanos , Imunidade Inata , Queratinócitos/citologia , Queratinócitos/imunologia , Queratinócitos/virologia , Fosforilação , RNA de Cadeia Dupla/imunologia , RNA Viral/imunologia , RNA Viral/metabolismo , Transdução de Sinais
13.
Duodecim ; 132(4): 349-54, 2016.
Artigo em Fi | MEDLINE | ID: mdl-27017787

RESUMO

Rheumatoid arthritis, inflammatory bowel diseases and psoriasis are examples of immune-mediated inflammatory diseases. They involve activation of a partly similar cytokine network that has an essential role in the disease pathogenesis. Biological drugs have been developed for the inhibition of single cytokines, and good therapeutic responses have been achieved by using them. For instance, TNF blockers are used in the treatment of several inflammatory diseases. The use of the blockers of certain other cytokines is more limited. Other important target molecules include certain interleukins. New bispecific antibodies enabling inhibition of the action of two distinct cytokines are currently undergoing clinical studies.


Assuntos
Artrite Reumatoide/imunologia , Produtos Biológicos/uso terapêutico , Terapia Biológica , Citocinas/antagonistas & inibidores , Citocinas/imunologia , Doenças Inflamatórias Intestinais/imunologia , Psoríase/imunologia , Artrite Reumatoide/tratamento farmacológico , Humanos , Doenças Inflamatórias Intestinais/tratamento farmacológico , Psoríase/tratamento farmacológico
14.
Proteomics ; 15(12): 2087-97, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25764225

RESUMO

Sendai virus (SeV) is a common respiratory pathogen in mice, rats, and hamsters. Host cell recognition of SeV is mediated by pathogen recognition receptors, which recognize viral components and induce intracellular signal transduction pathways that activate the antiviral innate immune response. Viruses use host proteins to control the activities of signaling proteins and their downstream targets, and one of the most important host protein modifications regulated by viral infection is phosphorylation. In this study, we used phosphoproteomics combined with bioinformatics to get a global view of the signaling pathways activated during SeV infection in human lung epithelial cells. We identified altogether 1347 phosphoproteins, and our data shows that SeV infection induces major changes in protein phosphorylation affecting the phosphorylation of almost one thousand host proteins. Bioinformatics analysis showed that SeV infection activates known pathways including MAPK signaling, as well as signaling pathways previously not linked to SeV infection including Rho family of GTPases, HIPPO signaling, and mammalian target of rapamycin (mTOR)-signaling pathway. Further, we performed functional studies with mTOR inhibitors and siRNA approach, which revealed that mTOR signaling is needed for both the host IFN response as well as viral protein synthesis in SeV-infected human lung epithelial cells.


Assuntos
Células Epiteliais/metabolismo , Neoplasias Pulmonares/metabolismo , Fosfoproteínas/metabolismo , Proteômica/métodos , Infecções por Respirovirus/metabolismo , Vírus Sendai/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Western Blotting , Biologia Computacional , Cricetinae , Células Epiteliais/citologia , Humanos , Interferons/metabolismo , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/virologia , Camundongos , Fosfoproteínas/genética , Fosforilação , Análise Serial de Proteínas , RNA Mensageiro/genética , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Infecções por Respirovirus/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Serina-Treonina Quinases TOR/genética , Células Tumorais Cultivadas
15.
Int J Cancer ; 137(7): 1775-83, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25821063

RESUMO

Metastatic melanoma is refractory to irradiation and chemotherapy, but amenable to immunological approaches such as immune-checkpoint-inhibiting antibodies or adoptive cell therapies. Oncolytic virus replication is an immunogenic phenomenon, and viruses can be armed with immunostimulatory molecules. Therefore, oncolytic immuno-virotherapy of malignant melanoma is an appealing approach, which was recently validated by a positive phase 3 trial. We investigated the potency of oncolytic adenovirus Ad5/3-D24-GMCSF on a panel of melanoma cell lines and animal models, and summarized the melanoma-specific human data from the Advanced Therapy Access Program (ATAP). The virus effectively eradicated human melanoma cells in vitro and subcutaneous SK-MEL-28 melanoma xenografts in nude mice when combined with low-dose cyclophosphamide. Furthermore, virally-expressed granulocyte-macrophage colony-stimulating factor (GM-CSF) stimulated the differentiation of human monocytes into macrophages. In contrast to human cells, RPMI 1846 hamster melanoma cells exhibited no response to oncolytic viruses and the chimeric 5/3 fiber failed to increase the efficacy of transduction, suggesting limited utility of the hamster model in the context of viruses with this capsid. In ATAP, treatments appeared safe and well-tolerated. Four out of nine melanoma patients treated were evaluable for possible therapy benefit with modified RECIST criteria: one patient had minor response, two had stable disease, and one had progressive disease. Two patients were alive at 559 and 2,149 days after treatment. Ad5/3-D24-GMCSF showed promising efficacy in preclinical studies and possible antitumor activity in melanoma patients refractory to other forms of therapy. This data supports continuing the clinical development of oncolytic adenoviruses for treatment of malignant melanoma.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Melanoma/terapia , Terapia Viral Oncolítica/métodos , Adenoviridae/genética , Animais , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Cricetinae , Ciclofosfamida/farmacologia , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Humanos , Macrófagos/patologia , Macrófagos/virologia , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/virologia , Camundongos , Camundongos Nus , Monócitos/patologia , Monócitos/virologia , Distribuição Aleatória , Ensaios Antitumorais Modelo de Xenoenxerto
16.
J Gen Virol ; 96(8): 2086-2091, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25934792

RESUMO

Non-structural protein NS1 of influenza A viruses interacts with cellular factors through its N-terminal RNA-binding, middle effector and C-terminal non-structured domains. NS1 attenuates antiviral responses in infected cells and thereby secures efficient virus replication. Some influenza strains express C-terminally truncated NS1 proteins due to nonsense mutations in the NS1 gene. To understand the role of the NS1 C-terminal region in regulation of antiviral responses, we engineered influenza viruses expressing C-terminally truncated NS1 proteins using A/WSN/33(H1N1) reverse genetics and tested them in human macrophages and in mice. We showed that a WSN virus expressing NS1 with a 28 aa deletion from its C terminus is a more powerful inducer of antiviral responses than the virus expressing full-length NS1, or one with a 10 aa truncation of NS1 in vitro. Thus, our findings suggest that the C-terminal region of NS1 is essential for regulation of antiviral responses. Moreover, viruses expressing truncated NS1 proteins could be good vaccine candidates.


Assuntos
Vírus da Influenza A Subtipo H1N1/imunologia , Influenza Humana/imunologia , Macrófagos/imunologia , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/imunologia , Motivos de Aminoácidos , Animais , Feminino , Humanos , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/fisiologia , Influenza Humana/virologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas não Estruturais Virais/genética , Replicação Viral
17.
Appl Environ Microbiol ; 81(8): 2939-49, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25681192

RESUMO

Amylosin, a heat-stable channel-forming non-ribosomally synthesized peptide toxin produced by strains of Bacillus amyloliquefaciens isolated from moisture-damaged buildings, is shown in this paper to have immunotoxic and cytotoxic effects on human cells as well as antagonistic effects on microbes. Human macrophages exposed to 50 ng of amylosin ml(-1) secreted high levels of cytokines interleukin-1ß (IL-1ß) and IL-18 within 2 h, indicating activation of the NLRP3 inflammasome, an integral part of the innate immune system. At the same exposure level, expression of IL-1ß and IL-18 mRNA increased. Amylosin caused dose-dependent potassium ion efflux from all tested mammalian cells (human monocytes and keratinocytes and porcine sperm cells) at 1 to 2 µM exposure. Amylosin also inhibited the motility of porcine sperm cells and depolarized the mitochondria of human keratinocytes. Amylosin may thus trigger the activation of the NLRP3 inflammasome and subsequently cytokine release by causing potassium efflux from exposed cells. The results of this study indicate that exposure to amylosin activates the innate immune system, which could offer an explanation for the inflammatory symptoms experienced by occupants of moisture-damaged buildings. In addition, the amylosin-producing B. amyloliquefaciens inhibited the growth of both prokaryotic and eukaryotic indoor microbes, and purified amylosin also had an antimicrobial effect. These antimicrobial effects could make amylosin producers dominant and therefore significant causal agents of health problems in some moisture-damaged sites.


Assuntos
Anti-Infecciosos/farmacologia , Bacillus/química , Bactérias/efeitos dos fármacos , Toxinas Bacterianas/toxicidade , Chaetomium/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Animais , Toxinas Bacterianas/imunologia , Humanos , Queratinócitos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Masculino , Potássio/metabolismo , Espermatozoides/efeitos dos fármacos , Suínos
18.
Mol Cell Proteomics ; 12(3): 749-63, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23292187

RESUMO

Monosodium urate (MSU) is an endogenous danger signal that is crystallized from uric acid released from injured cells. MSU is known to activate inflammatory response in macrophages but the molecular mechanisms involved have remained uncharacterized. Activated macrophages start to secrete proteins to activate immune response and to recruit other immune cells to the site of infection and/or tissue damage. Secretome characterization after activation of innate immune system is essential to unravel the details of early phases of defense responses. Here, we have analyzed the secretome of human primary macrophages stimulated with MSU using quantitative two-dimensional gel electrophoresis based proteomics as well as high-throughput qualitative GeLC-MS/MS approach combining protein separation by SDS-PAGE and protein identification by liquid chromatography-MS/MS. Both methods showed that MSU stimulation induced robust protein secretion from lipopolysaccharide-primed human macrophages. Bioinformatic analysis of the secretome data showed that MSU stimulation strongly activates unconventional, vesicle mediated protein secretion. The unconventionally secreted proteins included pro-inflammatory cytokines like IL-1ß and IL-18, interferon-induced proteins, and danger signal proteins. Also active forms of lysosomal proteases cathepsins were secreted on MSU stimulation, and cathepsin activity was essential for MSU-induced unconventional protein secretion. Additionally, proteins associated to phosphorylation events including Src family tyrosine kinases were increased in the secretome of MSU-stimulated cells. Our functional studies demonstrated that Src, Pyk2, and PI3 kinases act upstream of cathepsins to activate the overall protein secretion from macrophages. In conclusion, we provide the first comprehensive characterization of protein secretion pathways activated by MSU in human macrophages, and reveal a novel role for cathepsins and Src, Pyk2, PI3 kinases in the activation of unconventional protein secretion.


Assuntos
Catepsinas/metabolismo , Macrófagos/efeitos dos fármacos , Fosfotransferases/metabolismo , Proteoma/metabolismo , Ácido Úrico/farmacologia , Sequência de Aminoácidos , Antioxidantes/farmacologia , Catepsina B/antagonistas & inibidores , Catepsina B/metabolismo , Catepsinas/antagonistas & inibidores , Células Cultivadas , Quimiocinas/metabolismo , Cromatografia Líquida , Citocinas/metabolismo , Dipeptídeos/farmacologia , Eletroforese em Gel Bidimensional , Ativação Enzimática/efeitos dos fármacos , Quinase 2 de Adesão Focal/metabolismo , Humanos , Macrófagos/citologia , Macrófagos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteômica/métodos , Espectrometria de Massas em Tandem , Quinases da Família src/metabolismo
19.
J Proteome Res ; 13(5): 2468-77, 2014 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-24670152

RESUMO

Fungal infections (mycoses) are common diseases of varying severity that cause problems, especially to immunologically compromised people. Fungi express a variety of pathogen-associated molecular patterns on their surface including ß-glucans, which are important immunostimulatory components of fungal cell walls. During stimulatory conditions of infection and colonization, besides intensive intracellular response, human cells actively communicate on the intercellular level by secreting proteins and other biomolecules with several mechanisms. Vesicular secretion remains one of the most important paths for the proteins to exit the cell. Here, we have used high-throughput quantitative proteomics combined with bioinformatics to characterize and quantify vesicle-mediated protein release from ß-glucan-stimulated human macrophages differentiated in vitro from primary blood monocytes. We show that ß-glucan stimulation induces vesicle-mediated protein secretion. Proteomic study identified 540 distinct proteins from the vesicles, and the identified proteins show a proteomic signature characteristic for their cellular origin. Importantly, we identified several receptors, including cation-dependent mannose-6-phosphate receptor, macrophage scavenger receptor, and P2X7 receptor, that have not been identified from vesicles before. Proteomic data together with detailed pathway and network analysis showed that integrins and their cytoplasmic cargo proteins are highly abundant in extracellular vesicles released upon ß-glucan stimulation. In conclusion, the present data provides a solid basis for further studies on the functional role of vesicular protein secretion upon fungal infection.


Assuntos
Macrófagos/efeitos dos fármacos , Proteínas/metabolismo , Proteômica/métodos , Vesículas Secretórias/metabolismo , beta-Glucanas/farmacologia , Western Blotting , Diferenciação Celular , Células Cultivadas , Cromatografia Líquida , Humanos , Macrófagos/citologia , Macrófagos/metabolismo , Proteínas de Membrana/metabolismo , Microscopia Eletrônica , Monócitos/citologia , Proteoma/metabolismo , Vesículas Secretórias/ultraestrutura , Transdução de Sinais , Espectrometria de Massas em Tandem
20.
Antimicrob Agents Chemother ; 58(7): 3689-96, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24752266

RESUMO

The influenza pH1N1 virus caused a global flu pandemic in 2009 and continues manifestation as a seasonal virus. Better understanding of the virus-host cell interaction could result in development of better prevention and treatment options. Here we show that the Akt inhibitor MK2206 blocks influenza pH1N1 virus infection in vitro. In particular, at noncytotoxic concentrations, MK2206 alters Akt signaling and inhibits endocytic uptake of the virus. Interestingly, MK2206 is unable to inhibit H3N2, H7N9, and H5N1 viruses, indicating that pH1N1 evolved specific requirements for efficient infection. Thus, Akt signaling could be exploited further for development of better therapeutics against pH1N1 virus.


Assuntos
Compostos Heterocíclicos com 3 Anéis/farmacologia , Vírus da Influenza A Subtipo H1N1 , Influenza Humana/prevenção & controle , Proteína Oncogênica v-akt/antagonistas & inibidores , Inibidores de Proteases/farmacologia , Linhagem Celular , Citocinas/metabolismo , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Técnicas In Vitro , Influenza Humana/virologia , Dados de Sequência Molecular , Fosfoproteínas/metabolismo , RNA Interferente Pequeno/genética , Transfecção , Ensaio de Placa Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA