Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
mBio ; 12(5): e0219621, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34488451

RESUMO

Host shutoff in influenza A virus (IAV) infection is a key process contributing to viral takeover of the cellular machinery and resulting in the downregulation of host gene expression. Analysis of nascently transcribed RNA in a cellular model that allows the functional induction of NS1 demonstrates that NS1 suppresses host transcription. NS1 inhibits the expression of genes driven by RNA polymerase II as well as RNA polymerase I-driven promoters, but not by the noneukaryotic T7 polymerase. Additionally, transcriptional termination is deregulated in cells infected with wild-type IAV. The NS1 effector domain alone is able to mediate both effects, whereas NS1 mutant GLEWN184-188RFKRY (184-188) is not. Overexpression of CPSF30 counteracts NS1-mediated inhibition of RNA polymerase II-driven reporter gene expression, but knockdown of CPSF30 expression does not attenuate gene expression. Although NS1 is associated with nuclear chromatin, superresolution microscopy demonstrates that NS1 does not colocalize with genomic DNA. Moreover, NS1 mutants and NS1 fusion proteins, unable to associate with nuclear chromatin and displaying an altered subcellular distribution are still able to attenuate reporter gene expression. However, tethering NS1 artificially to the cytoskeleton results in the loss of reporter gene inhibition. A NS1 deficient in both native nuclear localization signals (NLS) is able to inhibit gene expression as effective as wild-type NS1 when a synthetic NLS relocates it to specific structures of the nucleus. Colocalization experiments and reporter gene cotransfection experiments with a NS1 fusion guiding it to nuclear speckles suggest that the presence of NS1 in nuclear speckles seems to be essential for host shutoff. IMPORTANCE We investigated the role of IAV nonstructural protein 1 NS1 in host gene shutoff-a central feature of IAV replication. We demonstrate that the effector domain of NS1 alone mediates host gene shutoff by inhibition of host transcription and by deregulation of the polyadenylation (polyA) signal-mediated 3' termination of host transcription. NS1 mutated in amino acids 184 to 188 fails to shut off host gene expression. Knockdown of CPSF30 does not result in transcriptional attenuation. By analyzing the subcellular localization of modified NS1 proteins and relating these data to their ability to inhibit reporter gene expression, we show for the first time that the presence of NS1 in granular structures of the nucleus-representing most likely nuclear speckles-seems to be essential to mediate host gene shutoff. Thus, our data present so far unknown insights into the molecular and spatial requirements needed for IAV-NS1-mediated host shutoff.


Assuntos
Núcleo Celular/virologia , Vírus da Influenza A Subtipo H7N7/metabolismo , Influenza Humana/genética , Influenza Humana/virologia , Transcrição Gênica , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Fator de Especificidade de Clivagem e Poliadenilação/genética , Fator de Especificidade de Clivagem e Poliadenilação/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza A Subtipo H7N7/genética , Influenza Humana/metabolismo , Domínios Proteicos , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Proteínas não Estruturais Virais/genética
2.
Cell Microbiol ; 19(7)2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28076660

RESUMO

Expression of the influenza A virus (IAV) nonstructural protein (NS1) results in the activation of c-Jun N-terminal kinase (JNK). Both NS1 and JNK are involved in apoptosis induction. To investigate their interrelationship, we stably expressed a tamoxifen inducible NS1 oestrogen receptor fusion-protein (NS1ERT) in mammalian cells. Upon tamoxifen stimulation, NS1ERT-expressing cells partially rescued the attenuated replication of NS1-deficient IAVs and also inhibited interferon up-regulation, confirming the functional competence of NS1ERT. Tamoxifen-induced NS1ERT created a cytopathic phenotype and led to the activation of JNK and apoptosis. Induction of NS1F103SERT mutant failed to activate JNK, but induced apoptosis, whereas the induction of NS1M106IERT led to JNK phosphorylation, but not apoptosis, indicating that JNK activation and apoptosis induction are not functionally linked. Further mutational analysis highlighted that apoptosis induction is a function of the C-terminal effector domain of NS1. Finally, IAVs encoding mutant NS1 revealed a modulating effect of NS1 on apoptosis induction in a genuine infection. With respect to apoptogenicity, an NS1 mutant virus that results in a super activation of JNK behaves similarly to the JNK nonactivating virus expressing NS1F103S, thus confirming that NS1-mediated JNK activation and apoptosis induction are also functionally independent from each other in vivo.


Assuntos
Apoptose/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Receptores de Estrogênio/genética , Proteínas Recombinantes de Fusão/genética , Proteínas não Estruturais Virais/genética , Células A549 , Animais , Linhagem Celular , Cães , Ativação Enzimática , Células HEK293 , Humanos , Vírus da Influenza A/metabolismo , Células Madin Darby de Rim Canino , Receptores de Estrogênio/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Tamoxifeno/metabolismo , Proteínas não Estruturais Virais/metabolismo
3.
J Virol ; 88(16): 8843-52, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24872593

RESUMO

UNLABELLED: A hallmark cell response to influenza A virus (IAV) infections is the phosphorylation and activation of c-jun N-terminal kinase (JNK). However, so far it is not fully clear which molecules are involved in the activation of JNK upon IAV infection. Here, we report that the transfection of influenza viral-RNA induces JNK in a retinoic acid-inducible gene I (RIG-I)-dependent manner. However, neither RIG-I-like receptors nor MyD88-dependent Toll-like receptors were found to be involved in the activation of JNK upon IAV infection. Viral JNK activation may be blocked by addition of cycloheximide and heat shock protein inhibitors during infection, suggesting that the expression of an IAV-encoded protein is responsible for JNK activation. Indeed, the overexpression of nonstructural protein 1 (NS1) of certain IAV subtypes activated JNK, whereas those of some other subtypes failed to activate JNK. Site-directed mutagenesis experiments using NS1 of the IAV H7N7, H5N1, and H3N2 subtypes identified the amino acid residue phenylalanine (F) at position 103 to be decisive for JNK activation. Cleavage- and polyadenylation-specific factor 30 (CPSF30), whose binding to NS1 is stabilized by the amino acids F103 and M106, is not involved in JNK activation. Conclusively, subtype-specific sequence variations in the IAV NS1 protein result in subtype-specific differences in JNK signaling upon IAV infection. IMPORTANCE: Influenza A virus (IAV) infection leads to the activation or modulation of multiple signaling pathways. Here, we demonstrate for the first time that the c-jun N-terminal kinase (JNK), a long-known stress-activated mitogen-activated protein (MAP) kinase, is activated by RIG-I when cells are treated with IAV RNA. However, at the same time, nonstructural protein 1 (NS1) of IAV has an intrinsic JNK-activating property that is dependent on IAV subtype-specific amino acid variations around position 103. Our findings identify two different and independent pathways that result in the activation of JNK in the course of an IAV infection.


Assuntos
Sequência de Aminoácidos/genética , Vírus da Influenza A/genética , Influenza Humana/genética , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Infecções por Orthomyxoviridae/genética , Proteínas não Estruturais Virais/genética , Animais , Linhagem Celular , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Cães , Células HEK293 , Humanos , Vírus da Influenza A/metabolismo , Influenza Humana/metabolismo , Influenza Humana/virologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Células Madin Darby de Rim Canino , Mutagênese Sítio-Dirigida/métodos , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Transdução de Sinais/genética , Tretinoína/metabolismo , Proteínas não Estruturais Virais/metabolismo
4.
PLoS One ; 9(2): e88924, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24586443

RESUMO

Our previous study presented evidence that the inflammation-related S100A9 gene is significantly upregulated in the brains of Alzheimer's disease (AD) animal models and human AD patients. In addition, experiments have shown that knockdown of S100A9 expression improves cognition function in AD model mice (Tg2576), and these animals exhibit reduced amyloid plaque burden. In this study, we established a new transgenic animal model of AD by crossbreeding the Tg2576 mouse with the S100A9 knockout (KO) mouse. We observed that S100A9KO/Tg2576 (KO/Tg) mice displayed an increased spatial reference memory in the Morris water maze task and Y-maze task as well as decreased amyloid beta peptide (Aß) neuropathology because of reduced levels of Aß, C-terminal fragments of amyloid precursor protein (APP-CT) and phosphorylated tau and increased expression of anti-inflammatory IL-10 and also decreased expression of inflammatory IL-6 and tumor neurosis factor (TNF)-α when compared with age-matched S100A9WT/Tg2576 (WT/Tg) mice. Overall, these results suggest that S100A9 is responsible for the neurodegeneration and cognitive deficits in Tg2576 mice. The mechanism of S100A9 is able to coincide with the inflammatory process. These findings indicate that knockout of S100A9 is a potential target for the pharmacological therapy of AD.


Assuntos
Calgranulina B/genética , Transtornos Cognitivos/genética , Transtornos da Memória/genética , Camundongos Knockout/genética , Doenças Neurodegenerativas/genética , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Calgranulina B/metabolismo , Transtornos Cognitivos/metabolismo , Modelos Animais de Doenças , Feminino , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Transtornos da Memória/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout/metabolismo , Camundongos Transgênicos/genética , Camundongos Transgênicos/metabolismo , Doenças Neurodegenerativas/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
5.
J Clin Invest ; 123(1): 206-14, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23202729

RESUMO

Influenza causes substantial morbidity and mortality, and highly pathogenic and drug-resistant strains are likely to emerge in the future. Protease-activated receptor 1 (PAR1) is a thrombin-activated receptor that contributes to inflammatory responses at mucosal surfaces. The role of PAR1 in pathogenesis of virus infections is unknown. Here, we demonstrate that PAR1 contributed to the deleterious inflammatory response after influenza virus infection in mice. Activating PAR1 by administering the agonist TFLLR-NH2 decreased survival and increased lung inflammation after influenza infection. Importantly, both administration of a PAR1 antagonist and PAR1 deficiency protected mice from infection with influenza A viruses (IAVs). Treatment with the PAR1 agonist did not alter survival of mice deficient in plasminogen (PLG), which suggests that PLG permits and/or interacts with a PAR1 function in this model. PAR1 antagonists are in human trials for other indications. Our findings suggest that PAR1 antagonism might be explored as a treatment for influenza, including that caused by highly pathogenic H5N1 and oseltamivir-resistant H1N1 viruses.


Assuntos
Vírus da Influenza A Subtipo H1N1/patogenicidade , Virus da Influenza A Subtipo H5N1/patogenicidade , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções por Orthomyxoviridae/imunologia , Receptor PAR-1/imunologia , Animais , Cães , Humanos , Influenza Humana/tratamento farmacológico , Influenza Humana/genética , Influenza Humana/imunologia , Camundongos , Camundongos Knockout , Células NIH 3T3 , Oligopeptídeos/farmacologia , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/patologia , Plasminogênio/genética , Plasminogênio/imunologia , Receptor PAR-1/agonistas , Receptor PAR-1/antagonistas & inibidores , Receptor PAR-1/genética
6.
Biol Chem ; 393(6): 525-34, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22628315

RESUMO

C-Jun N-terminal kinases (JNK) are activated in course of many viral infections. Here we analyzed the activity of JNK inhibitors on influenza A virus (IAV) amplification. Human lung epithelial cells were infected with either the highly pathogenic avian virus strain A/FPV/Bratislava/79 (H7N7) or the pandemic swine-origin influenza virus A/Hamburg/4/09 (H1N1v). The application of the JNK inhibitors SP600125 and AS601245 reduced IAV amplification by suppressing viral protein and RNA synthesis. Although AS601245 appeared to generally block the transcription of newly introduced genes, SP600125 specifically affected viral RNA synthesis. Overexpression of a dominant negative mutant of SEK/MKK4 and siRNA-mediated suppression of JNK2 expression confirmed that specific manipulation of the JNK pathway attenuates virus propagation. An IAV minigenome replication assay revealed that SP600125 did not directly affect the activity of the viral RNA polymerase complex but seems to suppress an anti-influenza nonstructural protein 1-mediated virus supportive function. Finally, when H7N7- or H1N1v-infected mice were treated with SP600125, the viral load is reduced in lungs of treated compared with untreated mice. Our data suggest that this class of ATP competitive inhibitors once optimized for antiviral action potentially represent novel drugs for antiviral intervention.


Assuntos
Antivirais/farmacologia , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/patogenicidade , Influenza Aviária/virologia , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Pandemias , Inibidores de Proteínas Quinases/farmacologia , Animais , Antivirais/química , Aves , Linhagem Celular , RNA Polimerases Dirigidas por DNA/metabolismo , Cães , Ativação Enzimática/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/patogenicidade , Vírus da Influenza A Subtipo H1N1/fisiologia , Vírus da Influenza A Subtipo H7N7/efeitos dos fármacos , Vírus da Influenza A Subtipo H7N7/genética , Vírus da Influenza A Subtipo H7N7/patogenicidade , Vírus da Influenza A Subtipo H7N7/fisiologia , Vírus da Influenza A/genética , Vírus da Influenza A/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/virologia , MAP Quinase Quinase 4/genética , Camundongos , Proteína Quinase 9 Ativada por Mitógeno/deficiência , Proteína Quinase 9 Ativada por Mitógeno/genética , Mutação , Biossíntese de Proteínas/efeitos dos fármacos , Inibidores de Proteínas Quinases/química , RNA Interferente Pequeno/genética , Transcrição Gênica/efeitos dos fármacos , Carga Viral/efeitos dos fármacos , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/efeitos dos fármacos
7.
FEBS Lett ; 586(2): 196-203, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22209981

RESUMO

Viral double-stranded RNA (dsRNA) and its synthetic analog polyI:C are recognized via multiple pathways and induce the expression of genes related to inflammation. In the present study, we demonstrated the polyI:C-induced gene expression of the damage associated molecular pattern (DAMP) molecules S100A8 and S100A9, while other S100 genes were not affected. Cycloheximide and Brefeldin A treatment revealed both the expression of S100A8 and S100A9 as secondary response genes and the involvement of polyI:C-induced cytokines herein. Several type I and type III interferons such as IFNß, IL-20, IL-24, and IFNλ/IL-29 were expressed in response to polyI:C, however, they failed to induce S100A8 and S100A9 gene expression. These data indicate the involvement of the danger molecule S100A8/A9 in the resistance against viruses.


Assuntos
Cicloeximida/farmacologia , Expressão Gênica , RNA de Cadeia Dupla/fisiologia , Proteínas S100/genética , Fatores de Transcrição/fisiologia , Imunidade Adaptativa/efeitos dos fármacos , Imunidade Adaptativa/genética , Imunidade Adaptativa/fisiologia , Brefeldina A/farmacologia , Calgranulina A/genética , Calgranulina A/metabolismo , Calgranulina B/genética , Calgranulina B/metabolismo , Células Cultivadas , Expressão Gênica/efeitos dos fármacos , Herpes Simples/genética , Herpes Simples/metabolismo , Herpes Simples/patologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiologia , Humanos , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Queratinócitos/patologia , Poli I-C/farmacologia , RNA de Cadeia Dupla/farmacologia , RNA Viral/genética , RNA Viral/fisiologia , Proteínas S100/metabolismo , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Regulação para Cima/efeitos dos fármacos
8.
Inflamm Allergy Drug Targets ; 10(3): 171-9, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21428909

RESUMO

Viral double-stranded RNA (dsRNA) and its synthetic analog poly (I:C) are recognized via multiple pathways and induce the expression of genes related to inflammation. In the present study, we demonstrate that poly (I:C) specifically induced the expression of matrix metallo-proteinase-9 (MMP-9) in HaCaT keratinocytes. Studies using specific pharmacological inhibitors revealed the involvement of NF-κB, p38 MAPK, and PI-3K signal transduction pathways in poly (I:C)-induced MMP-9 gene expression. MMP-9 gene induction was sensitive toward treatment with the macrolide antibiotic bafilomycin A1, a vacuolar H(+)-ATPase inhibitor, and with the lysosomotropic agent chloroquine. However, cycloheximide treatment only partially blocked poly (I:C)-induced MMP-9 gene expression. Although HaCaT keratinocytes produce a number of cytokines and chemokines in response to poly (I:C), stimulation experiments revealed that exclusively TNFα strongly promoted MMP-9 gene expression. During the antiviral response MMP-9 expression may be of importance for the tissue injury phase.


Assuntos
Herpes Simples/imunologia , Herpesvirus Humano 1/imunologia , Queratinócitos/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Pele/metabolismo , Linhagem Celular Transformada , Herpesvirus Humano 1/patogenicidade , Humanos , Queratinócitos/efeitos dos fármacos , Queratinócitos/imunologia , Queratinócitos/patologia , Queratinócitos/virologia , Macrolídeos/farmacologia , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/imunologia , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Poli I-C/imunologia , Poli I-C/metabolismo , RNA de Cadeia Dupla/imunologia , Transdução de Sinais , Pele/imunologia , Pele/patologia , Pele/virologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima/efeitos dos fármacos , ATPases Vacuolares Próton-Translocadoras/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
FEBS Lett ; 585(2): 440-6, 2011 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-21192933

RESUMO

S100A8/A9 promotes NADPH oxidase in HaCaT keratinocytes and subsequently increases NFκB activation, which plays important roles in the balance between epidermal growth and differentiation. S100A8/A9-positive HaCaT cells present with a significantly reduced rate of cell division and greater expression of two keratinocyte differentiation markers, involucrin and filaggrin, than control cells. S100A8/A9 mutants fail to enhance NFκB activation, TNFα-induced IL-8 gene expression and NFκB p65 phosphorylation, and S100A8/A9-positive cells demonstrate better cell survival in forced suspension culture than mutant cells. S100A8/A9 is induced in epithelial cells in response to stress. Therefore, S100A8/A9-mediated growth arrest could have implications for tissue remodeling and repair.


Assuntos
Calgranulina A/genética , Calgranulina B/genética , Diferenciação Celular , Proliferação de Células , Queratinócitos/citologia , Adaptação Fisiológica , Linhagem Celular , Células Epiteliais/citologia , Proteínas Filagrinas , Humanos , Queratinócitos/metabolismo , NF-kappa B/metabolismo
10.
Nat Med ; 16(6): 713-7, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20473308

RESUMO

Mechanisms linking innate immunity and autoimmune responses are poorly understood. Myeloid-related protein-8 (Mrp8) and Mrp14 are damage-associated molecular pattern molecules (DAMPs) highly upregulated in various autoimmune disorders. We show in a mouse autoimmune model that local Mrp8 and Mrp14 production is essential for the induction of autoreactive CD8+ T cells and the development of systemic autoimmunity. This effect is mediated via Toll-like receptor 4 (TLR4) signaling leading to increased interleukin-17 (IL-17) expression. Notably, expression of Mrp8 and Mrp14 was upregulated in cutaneous lupus erythematosus, and stimulation of CD8+ T cells from individuals with lupus erythematosus with MRP proteins resulted in an upregulation of IL-17, suggesting a key role for MRP8 and MRP14 for the development of autoreactive lymphocytes during human autoimmunity as well. These results demonstrate a link between local expression of DAMP molecules and the development of systemic autoimmunity.


Assuntos
Autoimunidade/imunologia , Linfócitos T CD8-Positivos/imunologia , Calgranulina A/imunologia , Calgranulina B/imunologia , Receptor 4 Toll-Like/imunologia , Transferência Adotiva , Animais , Autoimunidade/fisiologia , Ligante de CD40/imunologia , Linfócitos T CD8-Positivos/fisiologia , Calgranulina A/fisiologia , Calgranulina B/fisiologia , Humanos , Interleucina-17/imunologia , Interleucina-17/fisiologia , Lúpus Eritematoso Cutâneo/imunologia , Lúpus Eritematoso Cutâneo/fisiopatologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor 4 Toll-Like/fisiologia , Regulação para Cima/imunologia , Regulação para Cima/fisiologia
11.
PLoS Pathog ; 5(10): e1000639, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19876394

RESUMO

Neutrophils are the first line of defense at the site of an infection. They encounter and kill microbes intracellularly upon phagocytosis or extracellularly by degranulation of antimicrobial proteins and the release of Neutrophil Extracellular Traps (NETs). NETs were shown to ensnare and kill microbes. However, their complete protein composition and the antimicrobial mechanism are not well understood. Using a proteomic approach, we identified 24 NET-associated proteins. Quantitative analysis of these proteins and high resolution electron microscopy showed that NETs consist of modified nucleosomes and a stringent selection of other proteins. In contrast to previous results, we found several NET proteins that are cytoplasmic in unstimulated neutrophils. We demonstrated that of those proteins, the antimicrobial heterodimer calprotectin is released in NETs as the major antifungal component. Absence of calprotectin in NETs resulted in complete loss of antifungal activity in vitro. Analysis of three different Candida albicans in vivo infection models indicated that NET formation is a hitherto unrecognized route of calprotectin release. By comparing wild-type and calprotectin-deficient animals we found that calprotectin is crucial for the clearance of infection. Taken together, the present investigations confirmed the antifungal activity of calprotectin in vitro and, moreover, demonstrated that it contributes to effective host defense against C. albicans in vivo. We showed for the first time that a proportion of calprotectin is bound to NETs in vitro and in vivo.


Assuntos
Candida albicans/imunologia , Complexo Antígeno L1 Leucocitário/imunologia , Neutrófilos/imunologia , Abscesso Abdominal/imunologia , Abscesso Abdominal/microbiologia , Análise de Variância , Animais , Antifúngicos/química , Antifúngicos/metabolismo , Células Cultivadas , Estruturas Celulares/química , Estruturas Celulares/imunologia , Estruturas Celulares/ultraestrutura , Histonas/química , Histonas/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Imuno-Histoquímica , Complexo Antígeno L1 Leucocitário/química , Complexo Antígeno L1 Leucocitário/metabolismo , Pneumopatias Fúngicas/imunologia , Pneumopatias Fúngicas/microbiologia , Camundongos , Camundongos Knockout , Ativação de Neutrófilo
12.
Biochim Biophys Acta ; 1792(12): 1198-204, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19835955

RESUMO

Several reports have recently demonstrated a detrimental role of Toll-like receptors (TLR) in cerebral ischemia, while there is little information about the endogenous ligands which activate TLR-signaling. The myeloid related proteins-8 and-14 (Mrp8/S100A8; Mrp14/S100A9) have recently been characterized as endogenous TLR4-agonists, and thus may mediate TLR-activation in cerebral ischemia. Interestingly, not only TLR-mRNAs, but also Mrp8 and Mrp14 mRNA were found to be induced in mouse brain between 3 and 48 h after transient 1 h focal cerebral ischemia/reperfusion. Mrp-protein was expressed in the ischemic hemisphere, and co-labeled with CD11b-positive cells. To test the hypothesis that Mrp-signaling contributes to the postischemic brain damage, we subjected Mrp14-deficient mice, which also lack Mrp8 protein expression, to focal cerebral ischemia. Mrp14-deficient mice had significantly smaller lesion volumes when compared to wild-type littermates (130+/-16 mm(3) vs. 105+/-28 mm(3)) at 2 days after transient focal cerebral ischemia (1 h), less brain swelling, and a reduced macrophage/microglia cell count in the ischemic hemisphere. We conclude that upregulation and signaling of Mrp-8 and-14 contribute to neuroinflammation and the progression of ischemic damage.


Assuntos
Isquemia Encefálica/metabolismo , Calgranulina A/fisiologia , Calgranulina B/fisiologia , Sistema Nervoso Central/lesões , Inflamação/etiologia , Neurônios/patologia , Animais , Isquemia Encefálica/patologia , Sistema Nervoso Central/metabolismo , Perfilação da Expressão Gênica , Técnicas Imunoenzimáticas , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
J Biol Chem ; 283(46): 31776-84, 2008 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-18786929

RESUMO

The calcium binding S100A8/A9 complex (MRP8/14; calgranulin) is considered as an important proinflammatory mediator in acute and chronic inflammation and has recently gained attention as a molecular marker up-regulated in various human cancers. Here, we report that S100A8/A9 is expressed in breast cancer cell lines and is up-regulated by interleukin-1beta and tumor necrosis factor-alpha in SKBR3 and MCF-7 cells. We identified the phospholipid-binding protein annexin A6 as a potential S100A8/A9 binding protein by affinity chromatography. This finding was verified by Southwestern overlay experiments and by coimmunoprecipitation with the S100A8/A9-specific monoclonal antibody 27E10. Immunocytochemical experiments demonstrated that S100A8/A9 and annexin A6 colocalize in SKBR3 breast cancer cells predominantly in membranous structures. Upon calcium influx both S100A8/A9 and annexin A6 are exposed on the cell surface of SKBR3 cells. Subcellular fractionation studies suggested that after A23187 stimulation membrane association of S100A8/A9 is not enhanced. However, both S100A8/A9 and annexin A6 are exposed on the cell surface of SKBR3 cells upon calcium influx. Experiments with artificial liposomes indicated that S100A8/A9 is able to associate with membranes independently of both annexin A6 and independently of calcium. Finally, cell surface expression of S100A8/A9 could not be observed in A23187-treated A431 and HaCaT cells. Both cell lines are known to be devoid of annexin A6. Repression of annexin A6 expression by small interfering RNA in SKBR3 cells abolishes the cell surface exposition of S100A8/A9 upon calcium influx, suggesting that annexin A6 contributes to the calcium-dependent cell surface exposition of the membrane associated-S100A8/A9 complex.


Assuntos
Anexina A6/metabolismo , Cálcio/metabolismo , Calgranulina A/metabolismo , Calgranulina B/metabolismo , Calgranulina A/genética , Calgranulina B/genética , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Humanos , Metabolismo dos Lipídeos , Ligação Proteica
14.
J Exp Med ; 205(10): 2235-49, 2008 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-18809714

RESUMO

Accumulation of myeloid-derived suppressor cells (MDSCs) associated with inhibition of dendritic cell (DC) differentiation is one of the major immunological abnormalities in cancer and leads to suppression of antitumor immune responses. The molecular mechanism of this phenomenon remains unclear. We report here that STAT3-inducible up-regulation of the myeloid-related protein S100A9 enhances MDSC production in cancer. Mice lacking this protein mounted potent antitumor immune responses and rejected implanted tumors. This effect was reversed by administration of wild-type MDSCs from tumor-bearing mice to S100A9-null mice. Overexpression of S100A9 in cultured embryonic stem cells or transgenic mice inhibited the differentiation of DCs and macrophages and induced accumulation of MDSCs. This study demonstrates that tumor-induced up-regulation of S100A9 protein is critically important for accumulation of MDSCs and reveals a novel molecular mechanism of immunological abnormalities in cancer.


Assuntos
Calgranulina B/metabolismo , Diferenciação Celular/fisiologia , Células Dendríticas/fisiologia , Células Mieloides/fisiologia , Neoplasias/imunologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Calgranulina B/genética , Células Dendríticas/citologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Feminino , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Macrófagos/citologia , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células Mieloides/citologia , Células NIH 3T3 , Neoplasias/patologia , Fenótipo , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo
15.
J Cell Physiol ; 216(2): 558-67, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18452188

RESUMO

Leukocyte infiltration is an early and critical event in the development of acute pancreatitis. However, the mechanism of leukocyte transmigration into the pancreas and the function of leukocytes in initiating acute pancreatitis are still poorly understood. Here, we studied the role of S100A9 (MRP14), a calcium binding protein specifically released by polymorph nuclear leukocytes (PMN), in the course of acute experimental pancreatitis. Acute pancreatitis was induced by repeated supramaximal caerulein injections in S100A9 deficient or S100A9 wild-type mice. We then determined S100A9 expression, trypsinogen activation peptide (TAP) levels, serum amylase and lipase activities, and tissue myeloperoxidase (MPO) activity. Cell-cell contact dissociation was analyzed in vitro with biovolume measurements of isolated acini after incubation with purified S100A8/A9 heterodimers, and in vivo as measurement of Evans Blue extravasation after intravenous application of S100A8/A9. Pancreatitis induced increased levels of S100A9 in the pancreas. However, infiltration of leukocytes and MPO activity in the lungs and pancreas during acute pancreatitis was decreased in S100A9-deficient mice and associated with significantly lower serum amylase and lipase activities as well as reduced intrapancreatic TAP-levels. Incubation of isolated pancreatic acini with purified S100A8/A9-heterodimers resulted in a rapid dissociation of acinar cell-cell contacts which was highly calcium-dependent. Consistent with these findings, in vivo application of S100A8/A9 in mice was in itself sufficient to induce pancreatic cell-cell contract dissociation as indicated by Evans Blue extravasation. These data show that the degree of intrapancreatic trypsinogen activation is influenced by the extent of leukocyte infiltration into the pancreas which, in turn, depends on the presence of S100A9 that is secreted from PMN. S100A9 directly affects leukocyte tissue invasion and mediates cell contact dissociation via its calcium binding properties.


Assuntos
Calgranulina B/metabolismo , Junções Intercelulares/metabolismo , Leucócitos/imunologia , Pâncreas , Pancreatite/imunologia , Pancreatite/patologia , Animais , Biomarcadores/metabolismo , Cálcio/metabolismo , Calgranulina A , Calgranulina B/genética , Ceruletídeo/metabolismo , Ceruletídeo/toxicidade , Colecistocinina/metabolismo , Ativação Enzimática , Humanos , Leucócitos/citologia , Pulmão/citologia , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pâncreas/citologia , Pâncreas/imunologia , Pancreatite/induzido quimicamente , Proteínas S100/metabolismo , Tripsinogênio/metabolismo
16.
Science ; 319(5865): 962-5, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18276893

RESUMO

Bacterial infection often results in the formation of tissue abscesses, which represent the primary site of interaction between invading bacteria and the innate immune system. We identify the host protein calprotectin as a neutrophil-dependent factor expressed inside Staphylococcus aureus abscesses. Neutrophil-derived calprotectin inhibited S. aureus growth through chelation of nutrient Mn2+ and Zn2+: an activity that results in reprogramming of the bacterial transcriptome. The abscesses of mice lacking calprotectin were enriched in metal, and staphylococcal proliferation was enhanced in these metal-rich abscesses. These results demonstrate that calprotectin is a critical factor in the innate immune response to infection and define metal chelation as a strategy for inhibiting microbial growth inside abscessed tissue.


Assuntos
Abscesso/microbiologia , Quelantes/metabolismo , Complexo Antígeno L1 Leucocitário/metabolismo , Manganês/metabolismo , Neutrófilos/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/crescimento & desenvolvimento , Abscesso/imunologia , Abscesso/metabolismo , Animais , Cálcio/metabolismo , Quelantes/farmacologia , Dimerização , Perfilação da Expressão Gênica , Nefropatias/imunologia , Nefropatias/metabolismo , Nefropatias/microbiologia , Complexo Antígeno L1 Leucocitário/genética , Complexo Antígeno L1 Leucocitário/farmacologia , Abscesso Hepático/metabolismo , Abscesso Hepático/microbiologia , Abscesso Hepático/patologia , Espectrometria de Massas , Camundongos , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/genética , Zinco/metabolismo
17.
FEBS Lett ; 581(26): 5127-30, 2007 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-17936757

RESUMO

S100A8, S100A9 and S100A12 proteins are associated with inflammation and tissue remodelling, both processes known to be associated with high protease activity. Here, we report that homo-oligomeric forms of S100A8 and S100A9 are readily degraded by proteases, but that the preferred hetero-oligomeric S100A8/A9 complex displays a high resistance even against proteinase K degradation. S100A12 is not as protease resistant as the S100A8/A9 complex. Since specific functions have been assigned to the homo- and heterooligomeric forms of the S100A8 and A9 proteins, this finding may point to a post-translational level of regulation of the various functions of these proteins in inflammation and tissue remodelling.


Assuntos
Calgranulina A/metabolismo , Calgranulina B/metabolismo , Endopeptidase K/metabolismo , Animais , Calgranulina A/química , Calgranulina A/genética , Calgranulina B/química , Calgranulina B/genética , Endopeptidase K/química
18.
Nat Med ; 13(9): 1042-9, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17767165

RESUMO

To identify new components that regulate the inflammatory cascade during sepsis, we characterized the functions of myeloid-related protein-8 (Mrp8, S100A8) and myeloid-related protein-14 (Mrp14, S100A9), two abundant cytoplasmic proteins of phagocytes. We now demonstrate that mice lacking Mrp8-Mrp14 complexes are protected from endotoxin-induced lethal shock and Escherichia coli-induced abdominal sepsis. Both proteins are released during activation of phagocytes, and Mrp8-Mrp14 complexes amplify the endotoxin-triggered inflammatory responses of phagocytes. Mrp8 is the active component that induces intracellular translocation of myeloid differentiation primary response protein 88 and activation of interleukin-1 receptor-associated kinase-1 and nuclear factor-kappaB, resulting in elevated expression of tumor necrosis factor-alpha (TNF-alpha). Using phagocytes expressing a nonfunctional Toll-like receptor 4 (TLR4), HEK293 cells transfected with TLR4, CD14 and MD2, and by surface plasmon resonance studies in vitro, we demonstrate that Mrp8 specifically interacts with the TLR4-MD2 complex, thus representing an endogenous ligand of TLR4. Therefore Mrp8-Mrp14 complexes are new inflammatory components that amplify phagocyte activation during sepsis upstream of TNFalpha-dependent effects.


Assuntos
Calgranulina A/fisiologia , Calgranulina B/fisiologia , Endotoxinas/toxicidade , Choque Séptico/fisiopatologia , Receptor 4 Toll-Like/fisiologia , Animais , Calgranulina A/deficiência , Calgranulina B/genética , Linhagem Celular Tumoral , Células Cultivadas , Citometria de Fluxo , Humanos , Ligantes , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/fisiologia , Choque Séptico/genética , Fator de Necrose Tumoral alfa/genética
19.
J Invest Dermatol ; 127(8): 2001-11, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17429438

RESUMO

The calcium- and arachidonic acid (AA)-binding proteins S100A8 and S100A9 are involved in nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activation in phagocytes. They are specifically expressed in myeloid cells, and are also found in epithelial cells in various (patho)physiological conditions. We have investigated the consequences of S100A8/A9 overexpression in epithelial cell lines on reactive oxygen species (ROS) generation and downstream signaling. Epithelial carcinoma HeLa cells, which exclusively express Nox2, showed dramatically increased activation of NADPH oxidase by phorbol 12-myristate 13-acetate after S100A8/A9 gene transfection. HaCaT keratinocytes overexpressing S100A8/A9 showed enhanced, transient ROS generation in response to the calcium ionophore A23187 compared to mock-transfected cells. Polymerase chain reaction analysis revealed mRNA transcripts for Nox1, Nox2, and Nox5 in HaCaT keratinocytes. Detailed transfection studies confirmed that NADPH oxidase activities in Nox1- and Nox5-transfected HeLa cells were enhanced after S100A8/A9 gene complementation. Furthermore, mutational analysis revealed that AA binding and Thr113 phosphorylation are important for S100A8/A9-enhanced activation of NADPH oxidase. Nuclear factor-kappaB (NF-kappaB) activation and interleukin-8 mRNA levels were increased in S100A8/A9-HaCaT keratinocytes, consistent with the view that NF-kappaB is a redox-sensitive transcription factor. Because they are expressed in epithelia under specific conditions, S100A8 and S100A9 might be involved in skin pathogenesis by modulating aspects of downstream signaling.


Assuntos
Calgranulina A/fisiologia , Calgranulina B/fisiologia , Queratinócitos/metabolismo , NADPH Oxidases/metabolismo , NF-kappa B/metabolismo , Calcimicina/farmacologia , Calgranulina A/análise , Calgranulina B/análise , Células Cultivadas , Humanos , Interleucina-8/biossíntese , NADPH Oxidases/genética , Fosfoproteínas/genética , RNA Mensageiro/análise , Espécies Reativas de Oxigênio/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Transfecção , Fator de Necrose Tumoral alfa/farmacologia
20.
Blood ; 109(3): 1265-74, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17018861

RESUMO

Monocytes and macrophages may either promote or down-regulate inflammatory reactions depending on their state of activation. The effects of glucocorticoids (GCs), the most widely used immunosuppressive drugs, on monocytes are currently not well defined. By analyzing the GC-induced expression pattern in human monocytes by microarray technology, we identified for the first time GC-dependent regulation of 133 genes, including anti-inflammatory molecules such as adenosine A3 receptor, CD1d, and IL-1 receptor II. The results were independently confirmed by real-time polymerase chain reaction (PCR) and flow cytometry. Functional clustering of GC-regulated genes indicated induction of monocytic properties such as phagocytosis and motility as well as repression of adhesion, apoptosis, and oxidative burst. These predictions were confirmed by independent functional assays. GCs up-regulate fMLP receptors and specifically promote chemotaxis to this chemoattractant. Furthermore, GCs promote survival of an anti-inflammatory monocytic phenotype in inflammatory reactions, probably by inhibition of apoptosis because of oxidative stress. GCs limit tissue damage because of induction of antioxidative properties and high capacity for phagocytosis of proinflammatory agents. Thus, GC treatment did not cause a global suppression of monocytic effector functions but results in differentiation of a specific anti-inflammatory phenotype which seems to be actively involved in resolution of inflammatory reactions.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Inflamação/genética , Monócitos/efeitos dos fármacos , Apoptose/genética , Adesão Celular/genética , Movimento Celular/genética , Perfilação da Expressão Gênica , Humanos , Fagocitose/genética , Fenótipo , Explosão Respiratória/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA