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








Base de dados
Intervalo de ano de publicação
1.
Am J Pathol ; 194(5): 810-827, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38325553

RESUMO

Corneal nerve impairment contributes significantly to dry eye disease (DED) symptoms and is thought to be secondary to corneal epithelial damage. Transient receptor potential vanilloid-1 (TRPV1) channels abound in corneal nerve fibers and respond to inflammation-derived ligands, which increase in DED. TRPV1 overactivation promotes axonal degeneration in vitro, but whether it participates in DED-associated corneal nerve dysfunction is unknown. To explore this, DED was surgically induced in wild-type and TRPV1-knockout mice, which developed comparable corneal epithelial damage and reduced tear secretion. However, corneal mechanosensitivity decreased progressively only in wild-type DED mice. Sensitivity to capsaicin (TRPV1 agonist) increased in wild-type DED mice, and consistently, only this strain displayed DED-induced pain signs. Wild-type DED mice exhibited nerve degeneration throughout the corneal epithelium, whereas TRPV1-knockout DED mice only developed a reduction in the most superficial nerve endings that failed to propagate to the deeper subbasal corneal nerves. Pharmacologic TRPV1 blockade reproduced these findings in wild-type DED mice, whereas CD4+ T cells from both strains were equally pathogenic when transferred, ruling out a T-cell-mediated effect of TRPV1 deficiency. These data show that ocular desiccation triggers superficial corneal nerve damage in DED, but proximal propagation of axonal degeneration requires TRPV1 expression. Local inflammation sensitized TRPV1 channels, which increased ocular pain. Thus, ocular TRPV1 overactivation drives DED-associated corneal nerve impairment.


Assuntos
Lesões da Córnea , Síndromes do Olho Seco , Canais de Potencial de Receptor Transitório , Animais , Camundongos , Córnea/patologia , Lesões da Córnea/patologia , Síndromes do Olho Seco/metabolismo , Inflamação/patologia , Dor , Canais de Potencial de Receptor Transitório/farmacologia
2.
Front Immunol ; 13: 832306, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36091026

RESUMO

Neutrophils play major roles against bacteria and fungi infections not only due to their microbicide properties but also because they release mediators like Interleukin-1 beta (IL-1ß) that contribute to orchestrate the inflammatory response. This cytokine is a leaderless protein synthesized in the cytoplasm as a precursor (pro-IL-1ß) that is proteolytically processed to its active isoform and released from human neutrophils by secretory autophagy. In most myeloid cells, pro-IL-1ß is processed by caspase-1 upon inflammasome activation. Here we employed neutrophils from both healthy donors and patients with a gain-of-function (GOF) NLRP3-mutation to dissect IL-1ß processing in these cells. We found that although caspase-1 is required for IL-1ß secretion, it undergoes rapid inactivation, and instead, neutrophil serine proteases play a key role in pro-IL-1ß processing. Our findings bring to light distinctive features of the regulation of caspase-1 activity in human neutrophils and reveal new molecular mechanisms that control human neutrophil IL-1ß secretion.


Assuntos
Autofagia , Caspase 1 , Interleucina-1beta , Neutrófilos , Serina Proteases , Autofagia/genética , Autofagia/imunologia , Caspase 1/genética , Caspase 1/metabolismo , Humanos , Inflamassomos/genética , Inflamassomos/imunologia , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Neutrófilos/enzimologia , Neutrófilos/imunologia , Serina Endopeptidases/genética , Serina Endopeptidases/imunologia , Serina Proteases/genética , Serina Proteases/imunologia
3.
Immunology ; 161(2): 148-161, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32702135

RESUMO

Dry eye disease (DED) is a highly prevalent ocular surface disorder with neuroimmune pathophysiology. Tear hyperosmolarity (THO), a frequent finding in affected patients, is considered a key element in DED pathogenesis, yet existing animal models are based on subjecting the ocular surface to the more complex desiccating stress - decreased tear production and/or increased evaporation - instead of strict hyperosmolar stress. Here we characterized a murine model of THO that does not involve desiccating stress, thus allowing us to dissect the contribution of THO to DED. Our results showed that THO is sufficient to disrupt neuroimmune homeostasis of the ocular surface in mice, and thus reproduce many sub-clinical DED findings. THO activated nuclear factor-κB signalling in conjunctival epithelial cells and increased dendritic cell recruitment and maturation, leading to more activated (CD69+ ) and memory (CD62lo CD44hi) CD4+ T-cells in the eye-draining lymph nodes. Ultimately, THO impaired the development of ocular mucosal tolerance to a topical surrogate antigen in a chain of events that included epithelial nuclear factor-κB signalling and activation of transient receptor potential vanilloid 1 as the probable hypertonicity sensor. Also, THO reduced the density of corneal intraepithelial nerves and terminals, and sensitized the ocular surface to hypertonicity. Finally, the adoptive transfer of T-cells from THO mice to naïve recipients under mild desiccating stress favoured DED development, showing that THO is enough to trigger an actual pathogenic T-cell response. Our results altogether demonstrate that THO is a critical initiating factor in DED development.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Síndromes do Olho Seco/fisiopatologia , Fenômenos Fisiológicos Oculares , Lágrimas/metabolismo , Transferência Adotiva , Animais , Células Cultivadas , Olho , Homeostase , Humanos , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Neuroimunomodulação , Concentração Osmolar , Transdução de Sinais , Canais de Cátion TRPV/metabolismo , Lágrimas/química
4.
Front Immunol ; 11: 524180, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33692774

RESUMO

The type 1 TNF-α receptor (TNFR1) has a central role in initiating both pro-inflammatory and pro-apoptotic signaling cascades in neutrophils. Considering that TNFR1 signals Staphylococcus aureus protein A (SpA), the aim of this study was to explore the interaction of this bacterial surface protein with neutrophils and keratinocytes to underscore the signaling pathways that may determine the fate of these innate immune cells in the infected tissue during staphylococcal skin infections. Using human neutrophils cultured in vitro and isogenic staphylococcal strains expressing or not protein A, we demonstrated that SpA is a potent inducer of IL-8 in neutrophils and that the induction of this chemokine is dependent on the SpA-TNFR1 interaction and p38 activation. In addition to IL-8, protein A induced the expression of TNF-α and MIP-1α highlighting the importance of SpA in the amplification of the inflammatory response. Protein A contributed to reduce neutrophil mortality prolonging their lifespan upon the encounter with S. aureus. Signaling initiated by SpA modulated the type of neutrophil cell death in vitro and during skin and soft tissue infections (SSTI) in vivo triggering the apoptotic pathway instead of necrosis. Moreover, SpA induced pro-inflammatory cytokines in keratinocytes, modulating their survival in vitro and preventing the exacerbated necrosis and ulceration of the epithelium during SSTI in vivo. Taken together, these results highlight the importance of the inflammatory signaling induced by protein A in neutrophils and skin epithelial cells. The ability of protein A to modulate the neutrophil/epithelial cell death program in the skin is of clinical relevance considering that lysis of neutrophils and epithelial cells will promote an intense inflammatory response and contribute to tissue damage, a non-desirable feature of complicated SSTI.


Assuntos
Queratinócitos/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , Neutrófilos/imunologia , Proteína Estafilocócica A/imunologia , Staphylococcus aureus/imunologia , Citocinas/imunologia , Humanos , Queratinócitos/microbiologia , Neutrófilos/microbiologia , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia
5.
Front Immunol ; 10: 2374, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31681277

RESUMO

Fever is a hallmark of infections and inflammatory diseases, represented by an increase of 1-4°C in core body temperature. Fever-range hyperthermia (FRH) has been shown to increase neutrophil recruitment to local sites of infection. Here, we evaluated the impact of a short period (1 h) of FRH (STFRH) on pro-inflammatory and bactericidal human neutrophil functions. STFRH did not affect neutrophil spontaneous apoptosis but reverted the lipopolysaccharide (LPS)-induced anti-apoptotic effect compared with that under normothermic conditions. Furthermore, STFRH accelerated phorbol myristate acetate (PMA)-induced NETosis evaluated either by the nuclear DNA decondensation at 2 h post-stimulation or by the increase in extracellular DNA that colocalized with myeloperoxidase (MPO) at 4 h post-stimulation. Increased NETosis upon STFRH was associated with an increase in reactive oxygen species (ROS) production but not in autophagy levels. STFRH also increased NETosis in response to Pseudomonas aeruginosa challenge but moderately reduced its phagocytosis. However, these STFRH-induced effects did not influence the ability of neutrophils to kill bacteria after 4 h of co-culture. STFRH also significantly reduced neutrophil capacity to release the pro-inflammatory cytokines chemokine (C-X-C motif) ligand 8/interleukin 8 (CXCL8/IL-8) and IL-1ß in response to LPS and P. aeruginosa challenge. Altogether, these results indicate that a short and mild hyperthermal period is enough to modulate neutrophil responses to bacterial encounter. They also suggest that fever spikes during bacterial infections might lead neutrophils to trigger an emergency response promoting neutrophil extracellular trap (NET) formation to ensnare bacteria in order to wall off the infection and to reduce their release of pro-inflammatory cytokines in order to limit the inflammatory response.


Assuntos
Armadilhas Extracelulares/imunologia , Febre/imunologia , Interleucina-1beta/imunologia , Interleucina-8/imunologia , Neutrófilos/imunologia , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Armadilhas Extracelulares/microbiologia , Feminino , Febre/microbiologia , Febre/patologia , Humanos , Masculino , Neutrófilos/microbiologia , Neutrófilos/patologia , Infecções por Pseudomonas/patologia
6.
Mucosal Immunol ; 11(5): 1441-1453, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29867077

RESUMO

Immunological interdependence between the two eyes has been reported for the cornea and the retina but not for the ocular mucosal surface. Intriguingly, patients frequently report ocular surface-related symptoms in the other eye after unilateral ocular surgery. Here we show how unilateral eye injuries in mice affect the mucosal immune response of the opposite ocular surface. We report that, despite the lack of lymphatic cross-drainage, a neurogenic inflammatory reflex in the contralateral conjunctiva is sufficient to increase, first, epithelial nuclear factor kappa B signaling, then, dendritic cell maturation, and finally, expansion of effector, instead of regulatory, T cells in the draining lymph node, leading to disrupted ocular mucosal tolerance. We also show that damage to ocular surface nerves is required. Using pharmacological inhibitors and agonists, we identified transient receptor potential vanilloid 1 (TRPV1) channel as the receptor sensing tissue damage in the injured eye and substance P released in the opposite ocular surface as the effector of the sympathetic response. Finally, blocking either step prevented subsequent ocular allergic reactions in the opposite eye in a unilateral corneal alkali burn model. This study demonstrates that both ocular surfaces are immunologically linked and suggests potential therapeutic targets for intervention.


Assuntos
Olho/imunologia , Inflamação/imunologia , Mucosa/imunologia , Substância P/imunologia , Canais de Cátion TRPV/imunologia , Animais , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Hipersensibilidade/imunologia , Linfonodos/imunologia , Melanoma , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , NF-kappa B/imunologia , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia
7.
Front Immunol ; 9: 269, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29515581

RESUMO

Interleukin-1ß (IL-1ß), a major pro-inflammatory cytokine, is a leaderless cytosolic protein whose secretion does not follow the classical endoplasmic reticulum-to-Golgi pathway, and for which a canonical mechanism of secretion remains to be established. Neutrophils are essential players against bacterial and fungi infections. These cells are rapidly and massively recruited from the circulation into infected tissues and, beyond of displaying an impressive arsenal of toxic weapons effective to kill pathogens, are also an important source of IL-1ß in infectious conditions. Here, we analyzed if an unconventional secretory autophagy mechanism is involved in the exportation of IL-1ß by these cells. Our findings indicated that inhibition of autophagy with 3-methyladenine and Wortmannin markedly reduced IL-1ß secretion induced by LPS + ATP, as did the disruption of the autophagic flux with Bafilomycin A1 and E64d. These compounds did not noticeable affect neutrophil viability ruling out that the effects on IL-1ß secretion were due to cell death. Furthermore, VPS34IN-1, a specific autophagy inhibitor, was still able to reduce IL-1ß secretion when added after it was synthesized. Moreover, siRNA-mediated knockdown of ATG5 markedly reduced IL-1ß secretion in neutrophil-differentiated PLB985 cells. Upon LPS + ATP stimulation, IL-1ß was incorporated to an autophagic compartment, as was revealed by its colocalization with LC3B by confocal microscopy. Overlapping of IL-1ß-LC3B in a vesicular compartment peaked before IL-1ß increased in culture supernatants. On the other hand, stimulation of autophagy by cell starvation augmented the colocalization of IL-1ß and LC3B and then promoted neutrophil IL-1ß secretion. In addition, specific ELISAs indicated that although both IL-1ß and pro-IL-1ß are released to culture supernatants upon neutrophil stimulation, autophagy only promotes IL-1ß secretion. Furthermore, the serine proteases inhibitor AEBSF reduced IL-1ß secretion. Moreover, IL-1ß could be also found colocalizing with elastase, suggesting both some vesicles containing IL-1ß intersect azurophil granules content and that serine proteases also regulate IL-1ß secretion. Altogether, our findings indicate that an unconventional autophagy-mediated secretory pathway mediates IL-1ß secretion in human neutrophils.


Assuntos
Mediadores da Inflamação/metabolismo , Interleucina-1beta/metabolismo , Neutrófilos/imunologia , Adenina/análogos & derivados , Adenina/farmacologia , Trifosfato de Adenosina/imunologia , Autofagia/efeitos dos fármacos , Proteína 5 Relacionada à Autofagia/genética , Proteína 5 Relacionada à Autofagia/metabolismo , Linhagem Celular , Humanos , Lipopolissacarídeos/imunologia , Macrolídeos/farmacologia , Proteínas Associadas aos Microtúbulos/metabolismo , Transporte Proteico , RNA Interferente Pequeno/genética , Via Secretória , Serina Proteases/metabolismo , Wortmanina/farmacologia
8.
J Innate Immun ; 9(4): 387-402, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28467984

RESUMO

Tissue injury leads to the release of uric acid (UA). At high local concentrations, UA can form monosodium urate crystals (MSU). MSU and UA stimulate neutrophils to release extracellular traps (NET). Here, we investigated whether these NET could be involved in the development of inflammation by stimulating cytokine release by airway epithelial cells. We found that NET significantly increased the secretion of CXCL8/IL-8 and IL-6 by alveolar and bronchial epithelial cells. These effects were not observed when NETosis was inhibited by Diphenyleneiodonium, elastase inhibitor, or Cl-amidine. Similar findings were made with NET induced by cigarette smoke extract, suggesting that NET proinflammatory capacity is independent of the inducing stimulus. Furthermore, NET affected neither the viability and morphology of epithelial cells nor the barrier integrity of polarized cells. The epithelial stimulatory capacity of NET was not affected by degradation of DNA with micrococcal nuclease, treatment with heparin, or inhibition of the elastase immobilized to DNA, but it was significantly reduced by pretreatment with an anti-HMGB-1 blocking antibody. Altogether, our findings indicate that NET exert direct proinflammatory effects on airway epithelial cells that might contribute in vivo to the further recruitment of neutrophils and the perpetuation of inflammation upon lung tissue damage.


Assuntos
Brônquios/parasitologia , Armadilhas Extracelulares/metabolismo , Inflamação/imunologia , Interleucina-6/metabolismo , Neutrófilos/imunologia , Alvéolos Pulmonares/patologia , Mucosa Respiratória/imunologia , Anticorpos Bloqueadores/farmacologia , Células Cultivadas , Fumar Cigarros/efeitos adversos , Armadilhas Extracelulares/imunologia , Proteína HMGB1/imunologia , Humanos , Interleucina-8/metabolismo , Oniocompostos/farmacologia , Ornitina/análogos & derivados , Ornitina/farmacologia , Proteínas Secretadas Inibidoras de Proteinases/farmacologia , Mucosa Respiratória/patologia , Ácido Úrico/metabolismo
9.
Cell Death Dis ; 7(10): e2437, 2016 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-27787523

RESUMO

Neutrophils have the shortest lifespan among leukocytes and usually die via apoptosis, limiting their deleterious potential. However, this tightly regulated cell death program can be modulated by pathogen-associated molecular patterns (PAMPs), danger-associated molecular pattern (DAMPs), and inflammatory cytokines. We have previously reported that low pH, a hallmark of inflammatory processes and solid tumors, moderately delays neutrophil apoptosis. Here we show that fever-range hyperthermia accelerates the rate of neutrophil apoptosis at neutral pH but markedly increases neutrophil survival induced by low pH. Interestingly, an opposite effect was observed in lymphocytes; hyperthermia plus low pH prevents lymphocyte activation and promotes the death of lymphocytes and lymphoid cell lines. Analysis of the mechanisms through which hyperthermia plus low pH increased neutrophil survival revealed that hyperthermia further decreases cytosolic pH induced by extracellular acidosis. The fact that two Na+/H+ exchanger inhibitors, 5-(N-ethyl-N-isopropyl) amiloride (EIPA) and amiloride, reproduced the effects induced by hyperthermia suggested that it prolongs neutrophil survival by inhibiting the Na+/H+ antiporter. The neutrophil anti-apoptotic effect induced by PAMPs, DAMPs, and inflammatory cytokines usually leads to the preservation of the major neutrophil effector functions such as phagocytosis and reactive oxygen species (ROS) production. In contrast, our data revealed that the anti-apoptotic effect induced by low pH and hyperthermia induced a functional profile characterized by a low phagocytic activity, an impairment in ROS production and a high ability to suppress T-cell activation and to produce the angiogenic factors VEGF, IL-8, and the matrix metallopeptidase 9 (MMP-9). These results suggest that acting together fever and local acidosis might drive the differentiation of neutrophils into a profile able to promote both cancer progression and tissue repair during the late phase of inflammation, two processes that are strongly dependent on the local production of angiogenic factors by infiltrating immune cells.


Assuntos
Apoptose , Febre/patologia , Hipertermia Induzida , Neovascularização Fisiológica , Neutrófilos/patologia , Proliferação de Células , Humanos , Concentração de Íons de Hidrogênio , Fenótipo , Linfócitos T/metabolismo
10.
Exp Eye Res ; 151: 19-22, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27443502

RESUMO

Dry eye is a highly prevalent immune disorder characterized by a dysfunctional tear film and a Th1/Th17 T cell response at the ocular surface. The specificity of these pathogenic effector T cells remains to be determined, but auto-reactivity is considered likely. However, we have previously shown that ocular mucosal tolerance to an exogenous antigen is disrupted in a scopolamine-induced murine dry eye model and that it is actually responsible for disease progression. Here we report comparable findings in an entirely different murine model of dry eye that involves resection of the extraorbital lacrimal glands but no systemic muscarinic receptor blockade. Upon ocular instillation of ovalbumin, a delayed breakdown in mucosal tolerance to this antigen was observed in excised but not in sham-operated mice, which was mediated by interferon γ- and interleukin 17-producing antigen-specific T cells. Consistently, antigen-specific regulatory T cells were detectable in sham-operated but not in excised mice. As for other models of ocular surface disorders, epithelial activation of the NF-κB pathway by desiccating stress was determinant in the mucosal immune outcome. Underscoring the role of mucosal tolerance disruption in dry eye pathogenesis, its prevention by a topical NF-κB inhibitor led to reduced corneal damage in excised mice. Altogether these results show that surgically originated desiccating stress also initiates an abnormal Th1/Th17 T cell response to harmless exogenous antigens that reach the ocular surface. This event might actually contribute to corneal damage and challenges the conception of dry eye as a strictly autoimmune disease.


Assuntos
Síndromes do Olho Seco/diagnóstico , Tolerância Imunológica , Imunidade Celular , Aparelho Lacrimal/imunologia , Linfócitos T Reguladores/imunologia , Animais , Túnica Conjuntiva/imunologia , Túnica Conjuntiva/patologia , Córnea/imunologia , Córnea/patologia , Modelos Animais de Doenças , Progressão da Doença , Síndromes do Olho Seco/imunologia , Síndromes do Olho Seco/cirurgia , Aparelho Lacrimal/patologia , Aparelho Lacrimal/cirurgia , Camundongos , Mucosa/imunologia , Mucosa/patologia
11.
J Immunol ; 196(10): 4014-29, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-27084100

RESUMO

In this study, we demonstrate that the unlipidated (U) outer membrane protein (Omp) 19 from Brucella spp. is a competitive inhibitor of human cathepsin L. U-Omp19 inhibits lysosome cathepsins and APC-derived microsome activity in vitro and partially inhibits lysosomal cathepsin L activity within live APCs. Codelivery of U-Omp19 with the Ag can reduce intracellular Ag digestion and increases Ag half-life in dendritic cells (DCs). U-Omp19 retains the Ag in Lamp-2(+) compartments after its internalization and promotes a sustained expression of MHC class I/peptide complexes in the cell surface of DCs. Consequently, U-Omp19 enhances Ag cross-presentation by DCs to CD8(+) T cells. U-Omp19 s.c. delivery induces the recruitment of CD11c(+)CD8α(+) DCs and monocytes to lymph nodes whereas it partially limits in vivo Ag proteolysis inside DCs. Accordingly, this protein is able to induce CD8(+) T cell responses in vivo against codelivered Ag. Antitumor responses were elicited after U-Omp19 coadministration, increasing survival of mice in a murine melanoma challenge model. Collectively, these results indicate that a cysteine protease inhibitor from bacterial origin could be a suitable component of vaccine formulations against tumors.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Brucella/imunologia , Brucelose/imunologia , Linfócitos T CD8-Positivos/fisiologia , Vacinas Anticâncer/imunologia , Catepsinas/metabolismo , Células Dendríticas/imunologia , Imunoterapia/métodos , Lipoproteínas/metabolismo , Lisossomos/metabolismo , Melanoma/terapia , Animais , Antígenos de Neoplasias/imunologia , Apresentação Cruzada , Feminino , Ativação Linfocitária , Proteína 2 de Membrana Associada ao Lisossomo/metabolismo , Melanoma/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
12.
J Innate Immun ; 8(3): 284-98, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26967533

RESUMO

Interleukin 1 (IL-1) ß is a critical cytokine that orchestrates host defenses against Staphylococcus aureus and is crucial for the eradication of bacteria. The production and action of IL-1ß are regulated by multiple control pathways. Among them, IL-1RII (the type II IL-1 receptor) acts as a decoy receptor and has been shown to regulate the biological effects of IL-1ß. High levels of soluble IL-1RII are present in septic patients; however, the stimuli that regulate the expression and release of IL-1RII in pathological conditions are incompletely elucidated. In the present study, we demonstrated the ability of S. aureus and protein A to induce IL-1RII shedding in myeloid cells. The positive modulation of IL-1RII expression and cleavage was associated with the failure to detect IL-1ß in response to S. aureus both in vitro and in vivo, suggesting that the soluble form of the receptor could be masking the availability of IL-1ß. The absence of detectable IL-1ß was associated with low levels of inflammatory cytokines and chemokines known to be regulated by IL-1ß and with increased bacterial persistence. Modulation of decoy receptors during systemic S. aureus infection is proposed as a new strategy used by this bacterium to evade the immune response.


Assuntos
Interleucina-1beta/imunologia , Monócitos/imunologia , Neutrófilos/imunologia , Receptores Tipo II de Interleucina-1/metabolismo , Sepse/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Animais , Linhagem Celular , Regulação da Expressão Gênica , Humanos , Evasão da Resposta Imune , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Monócitos/microbiologia , Neutrófilos/microbiologia , Proteólise , Receptores Tipo II de Interleucina-1/genética , Proteína Estafilocócica A/imunologia
13.
Eur J Immunol ; 43(12): 3324-35, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23963575

RESUMO

Neutrophils are essential players in acute inflammatory responses. Upon stimulation, neutrophils activate NADPH oxidase, generating an array of reactive oxygen species (ROS). Interleukin-1 beta (IL-1ß) is a major proinflammatory cytokine synthesized as a precursor that has to be proteolytically processed to become biologically active. The role of ROS in IL-1ß processing is still controversial and has not been previously studied in neutrophils. We report here that IL-1ß processing in human neutrophils is dependent on caspase-1 and on the serine proteases elastase and/or proteinase 3. NADPH oxidase deficient neutrophils activated caspase-1 and did not exhibit differences in NALP3 expression, indicating that ROS are neither required for inflammasome activation nor for its priming, as has been reported for macrophages. Strikingly, ROS exerted opposite effects on the processing and secretion of IL-1ß; whereas ROS negatively controlled caspase-1 activity, as reported in mononuclear phagocytes, ROS were found to be necessary for the exportation of mature IL-1ß out of the cell, a role never previously described. The complex ROS-mediated regulation of neutrophil IL-1ß secretion might constitute a physiological mechanism to control IL-1ß-dependent inflammatory processes where neutrophils play a crucial role.


Assuntos
Inflamassomos/imunologia , Interleucina-1beta/imunologia , NADPH Oxidases/imunologia , Espécies Reativas de Oxigênio/imunologia , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , Caspase 1/genética , Caspase 1/imunologia , Caspase 1/metabolismo , Linhagem Celular , Feminino , Humanos , Inflamassomos/genética , Inflamassomos/metabolismo , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Masculino , Mieloblastina/genética , Mieloblastina/imunologia , Mieloblastina/metabolismo , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR , Espécies Reativas de Oxigênio/metabolismo
14.
J Immunol ; 184(11): 6386-95, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20421641

RESUMO

We previously demonstrated that extracellular bacterial DNA activates neutrophils through a CpG- and TLR9-independent mechanism. Biofilms are microbial communities enclosed in a polymeric matrix that play a critical role in the pathogenesis of many infectious diseases. Because extracellular DNA is a key component of biofilms of different bacterial species, the aim of this study was to determine whether it plays a role in the ability of biofilms to induce human neutrophil activation. We found that degradation of matrix extracellular DNA with DNase I markedly reduced the capacity of Pseudomonas aeruginosa biofilms to induce the release of the neutrophil proinflammatory cytokines IL-8 and IL-1beta (>75%); reduced the upregulation of neutrophil activation markers CD18, CD11b, and CD66b (p < 0.001); reduced the number of bacteria phagocytosed per neutrophil contacting the biofilm; and reduced the production of neutrophil extracellular traps. Consistent with these findings, we found that biofilms formed by the lasI rhlI P. aeruginosa mutant strain, exhibiting a very low content of matrix extracellular DNA, displayed a lower capacity to stimulate the release of proinflammatory cytokines by neutrophils, which was not decreased further by DNase I treatment. Together, our findings support that matrix extracellular DNA is a major proinflammatory component of P. aeruginosa biofilms.


Assuntos
Biofilmes/crescimento & desenvolvimento , DNA Bacteriano/imunologia , Ativação de Neutrófilo/imunologia , Neutrófilos/imunologia , Pseudomonas aeruginosa/fisiologia , Citocinas/biossíntese , Líquido Extracelular/química , Líquido Extracelular/microbiologia , Humanos , Microscopia Confocal , Neutrófilos/metabolismo
15.
Lab Invest ; 90(7): 1049-59, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20368700

RESUMO

Neutrophils are short-lived cells that rapidly undergo apoptosis. However, their survival can be regulated by signals from the environment. Flagellin, the primary component of the bacterial flagella, is known to induce neutrophil activation. In this study we examined the ability of flagellin to modulate neutrophil apoptosis. Neutrophils cultured for 12 and 24 h in the presence of flagellin from Salmonella typhimurium at concentrations found in pathological situations underwent a marked prevention of apoptosis. In contrast, Helicobacter pylori flagellin did not affect neutrophil survival, suggesting that Salmonella flagellin exerts the antiapoptotic effect by interacting with TLR5. The delaying in apoptosis mediated by Salmonella flagellin was coupled to higher expression levels of the antiapoptotic protein Mcl-1 and lower levels of activated caspase-3. Analysis of the signaling pathways indicated that Salmonella flagellin induced the activation of the p38 and ERK1/2 MAPK pathways as well as the PI3K/Akt pathway. Furthermore, it also stimulated IkappaBalpha degradation and the phosphorylation of the p65 subunit, suggesting that Salmonella flagellin also triggers NF-kappaB activation. Moreover, the pharmacological inhibition of ERK1/2 pathway and NF-kappaB activation partially prevented the antiapoptotic effects exerted by flagellin. Finally, the apoptotic delaying effect exerted by flagellin was also evidenced when neutrophils were cultured with whole heat-killed S. typhimurium. Both a wild-type and an aflagellate mutant S. typhimurium strain promoted neutrophil survival; however, when cultured in low bacteria/neutrophil ratios, the flagellate bacteria showed a higher capacity to inhibit neutrophil apoptosis, although both strains showed a similar ability to induce neutrophil activation. Taken together, our results indicate that flagellin delays neutrophil apoptosis by a mechanism partially dependent on the activation of ERK1/2 MAPK and NF-kappaB. The ability of flagellin to delay neutrophil apoptosis could contribute to perpetuate the inflammation during infections with flagellated bacteria.


Assuntos
Apoptose/efeitos dos fármacos , Flagelina/farmacologia , Neutrófilos/efeitos dos fármacos , Caspase 3/metabolismo , Sobrevivência Celular , Células Cultivadas , Flagelos/fisiologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína de Sequência 1 de Leucemia de Células Mieloides , NF-kappa B/metabolismo , Neutrófilos/enzimologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Infecções por Salmonella/imunologia , Salmonella typhimurium/fisiologia
16.
Mol Immunol ; 46(1): 37-44, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18701168

RESUMO

We have previously demonstrated that bacterial DNA induces neutrophil activation through a CpG- and TLR9-independent but MyD88-dependent-pathway. In this study we determined that GM-CSF enhances the activation of neutrophils by bacterial DNA. Granulocyte-macrophage colony-stimulating factor increased IL-8 and IL-1beta secretion, and CD11b-upregulation induced by single-stranded bacterial DNA. It also enhanced neutrophil IL-8 production induced by double-stranded bacterial DNA, methylated single-stranded DNA, plasmid DNA, and phosphorothioated-CpG and non-CpG-oligodeoxynucleotides. Together these observations indicated that GM-CSF enhances neutrophil responses triggered by bacterial DNA in a CpG-independent fashion. We also found that GM-CSF enhanced the activation of the MAPKs p38 and ERK1/2 induced by bacterial DNA. Moreover, the pharmacological inhibition of these pathways significantly diminished GM-CSF ability to increase neutrophil activation by bacterial DNA. Finally, we observed that GM-CSF was unable to increase the activation of MyD88(-/-) neutrophils by bacterial DNA. Our findings suggest that GM-CSF modulates the CpG-independent, MyD88-dependent neutrophil response to bacterial DNA, by increasing the activation of the MAPKs p38 and ERK1/2.


Assuntos
Ilhas de CpG/genética , DNA Bacteriano/farmacologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Ativação de Neutrófilo/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Animais , Antígeno CD11b/metabolismo , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Interleucina-8/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Neutrófilos/enzimologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
17.
Lab Invest ; 88(9): 926-37, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18626469

RESUMO

Bacterial DNA activates neutrophils through a CpG- and TLR9-independent mechanism. Neutrophil activation does not require DNA internalization, suggesting that it results from the interaction of bacterial DNA with a neutrophil surface receptor. The aim of this study was to characterize the interaction of bacterial DNA with the neutrophil surface. Bacterial DNA binding showed saturation and was inhibited by unlabeled DNA but not by other polyanions like yeast tRNA and poly-A. Resembling the conditions under which bacterial DNA triggers neutrophil activation, binding was not modified in the presence or absence of calcium, magnesium or serum. Treatment of neutrophils with proteases not only dramatically reduced bacterial DNA binding but also inhibited neutrophil activation induced by bacterial DNA. Experiments performed with DNA samples of different lengths obtained after digestion of bacterial DNA with DNase showed that only DNA fragments greater than approximately 170-180 nucleotides competed bacterial DNA binding and retained the ability to trigger cell activation. Treatment of neutrophils with chemoattractants or conventional agonists significantly increased bacterial DNA binding. Moreover, neutrophils that underwent transmigration through human endothelial cell monolayers even in the absence of chemoattractants, exhibited higher binding levels of bacterial DNA. Together, our findings provide evidence that binding of bacterial DNA to neutrophils is a receptor-mediated process that conditions the ability of DNA to trigger cell activation. We speculate that neutrophil recognition of bacterial DNA might be modulated by the balance of agonists present at inflammatory foci. This effect might be relevant in bacterial infections with a biofilm etiology, in which extracellular DNA could function as a potent neutrophil agonist.


Assuntos
DNA Bacteriano/metabolismo , Neutrófilos/metabolismo , Sequência de Bases , Biofilmes , Células Cultivadas , Primers do DNA , Escherichia coli/genética , Humanos
18.
J Immunol ; 177(6): 4037-46, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16951367

RESUMO

We have previously shown that bacterial DNA activates human neutrophils in a CpG-independent manner. In this study, we have characterized the signaling pathways involved in the activation mechanism. We found that p38 MAPK, ERK1/2, and JNK pathways, as well as the PI3K/Akt pathway, are activated by bacterial DNA. We also determined that bacterial DNA induces NF-kappaB and AP-1 activation. When analyzing the role of these pathways on neutrophil functions, we observed that up-regulation of CD11b triggered by bacterial DNA was decreased by pharmacological inhibitors of the p38 MAPK, ERK1/2, and JNK, whereas stimulation of IL-8 release was dependent on p38, ERK1/2, and NF-kappaB. Moreover, we found that IL-8 production was markedly enhanced by inhibition of JNK, suggesting that this pathway negatively modulates NF-kappaB-dependent transcription. We also observed that bacterial DNA stimulated IL-1R-associated kinase-1 kinase activity and its partial degradation. Finally, we determined that bacterial DNA stimulated CD11b up-regulation in TLR9(-/-) but not in MyD88(-/-) mouse neutrophils, supporting that bacterial DNA induces neutrophil activation through a TLR9-independent and MyD88-dependent pathway.


Assuntos
DNA Bacteriano/fisiologia , Sistema de Sinalização das MAP Quinases/imunologia , Neutrófilos/enzimologia , Neutrófilos/microbiologia , Animais , Células Cultivadas , Escherichia coli/genética , Escherichia coli/imunologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Neutrófilos/metabolismo
19.
J Immunol ; 172(5): 3196-204, 2004 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-14978127

RESUMO

It is widely appreciated that inflammatory responses in peripheral tissues are usually associated to the development of acidic microenvironments. Despite this, there are few studies aimed to analyze the effect of extracellular pH on immune cell functions. We analyzed the impact of acidosis on the behavior of dendritic cells (DCs) derived from murine bone marrow. We found that extracellular acidosis (pH 6.5) markedly stimulated the uptake of FITC-OVA, FITC-dextran, and HRP by DCs. In fact, to reach similar levels of endocytosis, DCs cultured at pH 7.3 required concentrations of Ag in the extracellular medium almost 10-fold higher compared with DCs cultured at pH 6.5. Not only the endocytic capacity of DCs was up-regulated by extracellular acidosis, but also the expression of CD11c, MHC class II, CD40, and CD86 as well as the acquisition of extracellular Ags by DCs for MHC class I-restricted presentation. Importantly, DCs pulsed with Ag under acidosis showed an improved efficacy to induce both specific CD8(+) CTLs and specific Ab responses in vivo. Our results suggest that extracellular acidosis improves the Ag-presenting capacity of DCs.


Assuntos
Apresentação de Antígeno , Antígenos/metabolismo , Células Dendríticas/imunologia , Espaço Extracelular/imunologia , Espaço Extracelular/metabolismo , Antígenos de Histocompatibilidade Classe I/imunologia , Concentração de Íons de Hidrogênio , Acidose/imunologia , Acidose/metabolismo , Animais , Formação de Anticorpos , Antígenos/biossíntese , Antígenos CD/biossíntese , Antígenos CD/metabolismo , Diferenciação Celular/imunologia , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Proteínas do Ovo/administração & dosagem , Proteínas do Ovo/imunologia , Proteínas do Ovo/metabolismo , Endocitose/imunologia , Feminino , Antígenos de Histocompatibilidade Classe I/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Ovalbumina/metabolismo , Fragmentos de Peptídeos , Linfócitos T Citotóxicos/imunologia , Regulação para Cima/imunologia
20.
Eur J Immunol ; 33(11): 3164-74, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14579285

RESUMO

Bacterial DNA stimulates macrophages, monocytes, B lymphocytes, NK cells, and dendritic cells in a CpG-dependent manner. In this work we demonstrate that bacterial DNA, but not mammalian DNA, induces human neutrophil activation as assessed by L-selectin shedding, CD11b upregulation, and stimulation of cellular shape change, IL-8 secretion, and cell migration. Induction of these responses is not dependent on the presence of unmethylated CpG motifs, as neutrophil stimulatory properties were neither modified by CpG-methylation of bacterial DNA nor reproduced by oligonucleotides bearing CpG motifs. We found that human neutrophils express Toll-like receptor (TLR) 9 mRNA. However, as expected for a CpG-independent mechanism, activation does not involve a TLR9-dependent signaling pathway; neutrophil stimulation was not prevented by immobilization of bacterial DNA or by wortmannin or chloroquine, two agents that inhibit TLR9 signaling. Of note, both single-stranded and double-stranded DNA were able to induce activation, suggesting that neutrophils might be activated by bacterial DNA at inflammatory foci even in the absence of conditions required to induce DNA denaturation. Our findings provide the first evidence that neutrophils might be alerted to the presence of invading bacteria through recognition of its DNA via a novel mechanism not involving CpG motifs.


Assuntos
Bactérias/imunologia , DNA Bacteriano/imunologia , Neutrófilos/imunologia , Movimento Celular/imunologia , Ilhas de CpG/imunologia , Humanos , Antígeno de Macrófago 1/metabolismo , Glicoproteínas de Membrana/metabolismo , Neutrófilos/metabolismo , Oligodesoxirribonucleotídeos/imunologia , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/fisiologia , Receptor Toll-Like 9 , Receptores Toll-Like
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA