Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mucosal Immunol ; 17(3): 346-358, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38447907

RESUMO

OM-85 is a bacterial lysate used in clinical practice to reduce duration and frequency of recurrent respiratory tract infections. Whereas knowledge of its regulatory effects in vivo has substantially advanced, the mechanisms of OM-85 sensing remain inadequately addressed. Here, we show that the immune response to OM-85 in the mouse is largely mediated by myeloid immune cells through Toll-like receptor (TLR) 4 in vitro and in vivo. Instead, in human immune cells, TLR2 and TLR4 orchestrate the response to OM-85, which binds to both receptors as shown by surface plasmon resonance assay. Ribonucleic acid-sequencing analyses of human monocyte-derived dendritic cells reveal that OM-85 triggers a pro-inflammatory signature and a unique gene set, which is not induced by canonical agonists of TLR2 or TLR4 and comprises tolerogenic genes. A largely overlapping TLR2/4-dependent gene signature was observed in individual subsets of primary human airway myeloid cells, highlighting the robust effects of OM-85. Collectively, our results suggest caution should be taken when relating murine studies on bacterial lysates to humans. Furthermore, our data shed light on how a standardized bacterial lysate shapes the response through TLR2 and TLR4, which are crucial for immune response, trained immunity, and tolerance.


Assuntos
Imunomodulação , Células Mieloides , Receptor 2 Toll-Like , Receptor 4 Toll-Like , Humanos , Receptor 2 Toll-Like/metabolismo , Receptor 2 Toll-Like/genética , Camundongos , Animais , Receptor 4 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Células Mieloides/imunologia , Células Mieloides/metabolismo , Células Dendríticas/imunologia , Transcriptoma , Células Cultivadas , Camundongos Knockout , Regulação da Expressão Gênica , Lisados Bacterianos
2.
J Allergy Clin Immunol ; 153(5): 1330-1343, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38369029

RESUMO

BACKGROUND: The development of atopic dermatitis (AD) drugs is challenged by many disease phenotypes and trial design options, which are hard to explore experimentally. OBJECTIVE: We aimed to optimize AD trial design using simulations. METHODS: We constructed a quantitative systems pharmacology model of AD and standard of care (SoC) treatments and generated a phenotypically diverse virtual population whose parameter distribution was derived from known relationships between AD biomarkers and disease severity and calibrated using disease severity evolution under SoC regimens. RESULTS: We applied this workflow to the immunomodulator OM-85, currently being investigated for its potential use in AD, and calibrated the investigational treatment model with the efficacy profile of an existing trial (thereby enriching it with plausible marker levels and dynamics). We assessed the sensitivity of trial outcomes to trial protocol and found that for this particular example the choice of end point is more important than the choice of dosing regimen and patient selection by model-based responder enrichment could increase the expected effect size. A global sensitivity analysis revealed that only a limited subset of baseline biomarkers is needed to predict the drug response of the full virtual population. CONCLUSIONS: This AD quantitative systems pharmacology workflow built around knowledge of marker-severity relationships as well as SoC efficacy can be tailored to specific development cases to optimize several trial protocol parameters and biomarker stratification and therefore has promise to become a powerful model-informed AD drug development and personalized medicine tool.


Assuntos
Biomarcadores , Ensaios Clínicos como Assunto , Dermatite Atópica , Dermatite Atópica/tratamento farmacológico , Humanos , Farmacologia em Rede , Fluxo de Trabalho , Fatores Imunológicos/uso terapêutico , Fatores Imunológicos/farmacologia , Simulação por Computador , Projetos de Pesquisa , Índice de Gravidade de Doença
3.
Front Immunol ; 13: 867022, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35603159

RESUMO

Respiratory syncytial virus (RSV) is a seasonal pathogen responsible for the highest percentage of viral bronchiolitis in pediatric patients. There are currently no vaccine available and therapeutic methods to mitigate the severity of RSV bronchiolitis are limited. OM-85, an oral standardized bacterial lysate isolated from human respiratory strains and widely used to prevent recurrent infections and/or exacerbations in populations at risk, has been shown to be effective and safe in children and adults. Here, we demonstrate that airway administration of OM-85 in Balb/c mice prior to infection prevents RSV-induced disease, resulting in inhibition of viral replication associated with less perivascular and peribronchial inflammation in the lungs. These protective effects are dose and time-dependent with complete protection using 1mg dose of OM-85 only four times intranasally. Mechanistic insights using this topical route in the airways revealed increased alveolar macrophages, a selective set of tolerogenic DCs, Treg and Th1 expansion in the lung, even in the absence of infection, contributing to a better Th1/Th2 balance and preventing ILC2 recruitment in the airways and associated inflammatory sequelae. OM-85 preventive treatment also improved antiviral response by increasing IFNß and its responsive genes in the lung. In vitro, OM-85 protects against RSV infection in a type I interferon pathway. Our animal model data suggest that intranasal use of OM-85 should be considered as a potential prophylactic product to prevent RSV bronchiolitis once human studies confirm these findings.


Assuntos
Bronquiolite Viral , Infecções por Vírus Respiratório Sincicial , Vírus Sincicial Respiratório Humano , Animais , Extratos Celulares , Criança , Humanos , Imunidade Inata , Linfócitos , Camundongos , Camundongos Endogâmicos BALB C
4.
Nat Commun ; 13(1): 1980, 2022 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-35418135

RESUMO

Respiratory disease trials are profoundly affected by non-pharmaceutical interventions (NPIs) against COVID-19 because they perturb existing regular patterns of all seasonal viral epidemics. To address trial design with such uncertainty, we developed an epidemiological model of respiratory tract infection (RTI) coupled to a mechanistic description of viral RTI episodes. We explored the impact of reduced viral transmission (mimicking NPIs) using a virtual population and in silico trials for the bacterial lysate OM-85 as prophylaxis for RTI. Ratio-based efficacy metrics are only impacted under strict lockdown whereas absolute benefit already is with intermediate NPIs (eg. mask-wearing). Consequently, despite NPI, trials may meet their relative efficacy endpoints (provided recruitment hurdles can be overcome) but are difficult to assess with respect to clinical relevance. These results advocate to report a variety of metrics for benefit assessment, to use adaptive trial design and adapted statistical analyses. They also question eligibility criteria misaligned with the actual disease burden.


Assuntos
COVID-19 , Transtornos Respiratórios , Infecções Respiratórias , Viroses , COVID-19/prevenção & controle , Ensaios Clínicos como Assunto , Controle de Doenças Transmissíveis/métodos , Humanos , Infecções Respiratórias/epidemiologia , SARS-CoV-2 , Viroses/epidemiologia
5.
J Allergy Clin Immunol ; 149(3): 943-956, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34560105

RESUMO

BACKGROUND: Microbial interventions against allergic asthma have robust epidemiologic underpinnings and the potential to recalibrate disease-inducing immune responses. Oral administration of OM-85, a standardized lysate of human airways bacteria, is widely used empirically to prevent respiratory infections and a clinical trial is testing its ability to prevent asthma in high-risk children. We previously showed that intranasal administration of microbial products from farm environments abrogates experimental allergic asthma. OBJECTIVES: We sought to investigate whether direct administration of OM-85 to the airway compartment protects against experimental allergic asthma; and to identify protective cellular and molecular mechanisms activated through this natural route. METHODS: Different strains of mice sensitized and challenged with ovalbumin or Alternaria received OM-85 intranasally, and cardinal cellular and molecular asthma phenotypes were measured. Airway transfer experiments assessed whether OM-85-treated dendritic cells protect allergen-sensitized, OM-85-naive mice against asthma. RESULTS: Airway OM-85 administration suppressed allergic asthma in all models acting on multiple innate and adaptive immune targets: the airway epithelium/IL-33/ILC2 axis, lung allergen-induced type 2 responses, and dendritic cells whose Myd88/Trif-dependent tolerogenic reprogramming was sufficient to transfer OM-85-induced asthma protection. CONCLUSIONS: We provide the first demonstration that administering a standardized bacterial lysate to the airway compartment protects from experimental allergic asthma by engaging multiple immune pathways. Because protection required a cumulative dose 27- to 46-fold lower than the one reportedly active through the oral route, the efficacy of intranasal OM-85 administration may reflect its direct access to the airway mucosal networks controlling the initiation and development of allergic asthma.


Assuntos
Asma , Interleucina-33 , Alérgenos , Animais , Extratos Celulares , Células Dendríticas , Modelos Animais de Doenças , Epitélio , Humanos , Imunidade Inata , Pulmão , Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina
6.
Ital J Pediatr ; 44(1): 112, 2018 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-30257691

RESUMO

BACKGROUND: Viral respiratory infections may promote bacterial super-infection decreasing the host immune response efficiency. However, using a mice model we recently demonstrated that preventive treatment with the bacterial extract OM-85 reduces the susceptibility to a secondary Streptococcus (S.) pneumoniae infection after influenza virus (I.V.) challenge. METHODS: To better characterize the efficacy of OM-85 against S. pneumoniae super-infection, a post-hoc analysis was conducted, comparing efficacy (survival) and morbidity signs (clinical score, body temperature and weight loss) in the OM-85 and the control (BLANC) groups of mice after: a) I.V. infection; b) primary S. pneumoniae infection and c) post-I.V. S. pneumoniae super-infection. RESULTS: After a sublethal I.V. dose, all mice stayed alive at day 5 and no differences in morbidity signs were detected between the OM-85 and the BLANC groups. However, OM-85 pretreatment led to a significantly reduction of the viral load in the lung on day 5 post viral infection and, on day 10, reduced neutrophilic inflammation while increasing influenza-specific CD8 + T-cell proportion in the airways. Conversely to viral infection, exposure to S. pneumoniae induced a dramatic reduction of survival, with no mice surviving on day 3 post infection in the BLANC group, whereas a partial protective effect was observed in OM-85 pre-treated mice (20% of mice surviving at day 3, and 10% at day 4 and 5). The morbidity data substantiated the survival results. Interestingly, in the "super-infection" study, when mice were exposed to a sublethal I.V. dose followed by a secondary S. pneumoniae infection, all mice died by day 4 in the BLANC group. In contrast, in the OM-85 treated group, the survival rate was 70% at day 4 and still 50% at day 5, with positive effects on the clinical scores and on the body temperature already detectable at days 1 and 2. CONCLUSIONS: The efficacy of OM-85 pre-treatment against S. pneumoniae super-infection reflects a strong and immediate immune reaction from the host, an event that can be explained in part by a "non-specific" activation of the immune system, a positive "immune effect" of the general OM-85- induced immune response against I.V.


Assuntos
Adjuvantes Imunológicos/farmacologia , Extratos Celulares/farmacologia , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções Pneumocócicas/prevenção & controle , Superinfecção/prevenção & controle , Imunidade Adaptativa , Animais , Líquido da Lavagem Broncoalveolar/citologia , Linfócitos T CD8-Positivos/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Pulmão/virologia , Camundongos Endogâmicos BALB C , Neutrófilos/metabolismo , Streptococcus pneumoniae/imunologia , Carga Viral/efeitos dos fármacos
7.
J Clin Invest ; 128(11): 4856-4869, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30153109

RESUMO

Chronic allergic inflammatory diseases are a major cause of morbidity, with allergic asthma alone affecting over 300 million people worldwide. Epidemiological studies demonstrate that environmental stimuli are associated with either the promotion or prevention of disease. Major reductions in asthma prevalence are documented in European and US farming communities. Protection is associated with exposure of mothers during pregnancy to microbial breakdown products present in farm dusts and unprocessed foods and enhancement of innate immune competence in the children. We sought to develop a scientific rationale for progressing these findings toward clinical application for primary disease prevention. Treatment of pregnant mice with a defined, clinically approved immune modulator was shown to markedly reduce susceptibility of their offspring to development of the hallmark clinical features of allergic airway inflammatory disease. Mechanistically, offspring displayed enhanced dendritic cell-dependent airway mucosal immune surveillance function, which resulted in more efficient generation of mucosal-homing regulatory T cells in response to local inflammatory challenge. We provide evidence that the principal target for maternal treatment effects was the fetal dendritic cell progenitor compartment, equipping the offspring for accelerated functional maturation of the airway mucosal dendritic cell network following birth. These data provide proof of concept supporting the rationale for developing transplacental immune reprogramming approaches for primary disease prevention.


Assuntos
Asma/imunologia , Bactérias/imunologia , Células Dendríticas/imunologia , Imunidade Materno-Adquirida , Placenta/imunologia , Linfócitos T Reguladores/imunologia , Animais , Asma/patologia , Asma/prevenção & controle , Células Dendríticas/patologia , Feminino , Imunidade nas Mucosas , Inflamação/imunologia , Inflamação/patologia , Inflamação/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Placenta/patologia , Gravidez , Ratos , Ratos Sprague-Dawley , Linfócitos T Reguladores/patologia
8.
PLoS One ; 12(11): e0188010, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29182620

RESUMO

BACKGROUND: Bronchial epithelial cells (BEC) are primary target for Rhinovirus infection through attaching to cell membrane proteins. OM-85, a bacterial extract, improves recovery of asthma and COPD patients after viral infections, but only part of the mechanism was addressed, by focusing on defined immune cells. OBJECTIVE: We therefore determined the effect of OM-85 on isolated primary human BEC of controls (n = 8), asthma patients (n = 10) and COPD patients (n = 9). METHODS: BEC were treated with OM-85 alone (24 hours) or infected with Rhinovirus. BEC survival was monitored by manual cell counting and Rhinovirus replication by lytic activity. Immuno-blotting and ELISA were used to determine the expression of Rhinovirus interacting proteins: intracellular adhesion molecule (ICAM), major histocompatibility complex class II (MHC-2), complement component C1q receptor (C1q-R), inducible T-Cell co-stimulator (ICOS), its ligand ICOSL, and myeloid differentiation primary response gene 88 (Myd88); as well as for signal transducers Erk1/2, p38, JNK mitogen activated protein kinases MAPK), and cAMP. RESULTS: OM-85 significantly reduced Rhinovirus-induced BEC death and virus replication. OM-85 significantly increased the expression of virus interacting proteins C1q-R and ß-defensin in all 3 probes and groups, which was prevented by either Erk1/2 MAPK or cAMP inhibition. In addition, OM-85 significantly reduced Rhinovirus induced expression of ICAM1 involving p38 MAPK. In BEC OM-85 had no significant effect on the expression of ICOS, ICOSL and MHC-2 membrane proteins nor on the adaptor protein MyD88. CONCLUSION: The OM-85-induced increased of C1q-R and ß-defensin, both important for antigen presentation and phagocytosis, supports its activity in host cell's defence against Rhinovirus infection.


Assuntos
Brônquios/metabolismo , Extratos Celulares/farmacologia , AMP Cíclico/metabolismo , Sistema de Sinalização das MAP Quinases , Rhinovirus/efeitos dos fármacos , Proteínas Virais/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Brônquios/citologia , Estudos de Casos e Controles , Células Cultivadas , Células Epiteliais/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Doença Pulmonar Obstrutiva Crônica/metabolismo , Doença Pulmonar Obstrutiva Crônica/patologia , Rhinovirus/metabolismo
9.
J Diabetes Complications ; 31(10): 1481-1490, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28847447

RESUMO

AIM: Calcium dobesilate (CaD) is beneficial in early stages of diabetic retinopathy (DR), but its mechanisms of action remains to be elucidated. The aim was to investigate the effect of CaD on proinflammatory cytokines and oxidative stress. METHODS: db/db mice were randomly assigned to daily oral treatment with CaD (200mg/kg/day) or vehicle for 15days. Biomarkers of oxidative stress (dihydroethidium, malondialdehyde), NF-κB, and proinflammatory cytokines (IL-1ß, IL-6, IL-8, TNF-α, MCP-1) were examined in the retina by immunohistochemical analysis. Cultures of human retinal endothelial cells (HRECs) were used for complementary experiments. RESULTS: CaD significantly reduced the biomarkers of oxidative stress in the retina of db/db mice. In addition, CaD prevented the increase of NF-κB, IL-6, IL-8, TNF-α and MCP-1 induced by diabetes. CaD inhibited the activation of NF-kß induced by IL-1ß by preventing IKKB-α phosphorylation in HRECs and reduced the upregulation of IL-6 and IL-18 induced by TNF-α in a dose-dependent manner. CONCLUSION: Our results suggest that antioxidant and antiinflammatory effects are crucial in accounting for the effectiveness of CaD for treating DR.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Dobesilato de Cálcio/uso terapêutico , Retinopatia Diabética/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Retina/efeitos dos fármacos , Retinite/prevenção & controle , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Biomarcadores/metabolismo , Dobesilato de Cálcio/farmacologia , Células Cultivadas , Cruzamentos Genéticos , Citocinas/antagonistas & inibidores , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/tratamento farmacológico , Retinopatia Diabética/imunologia , Retinopatia Diabética/metabolismo , Retinopatia Diabética/patologia , Humanos , Quinase I-kappa B/metabolismo , Masculino , Camundongos Mutantes , Camundongos Transgênicos , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Distribuição Aleatória , Retina/imunologia , Retina/metabolismo , Retina/patologia , Retinite/complicações , Retinite/imunologia , Retinite/metabolismo
10.
Curr Eye Res ; 42(9): 1273-1286, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28574750

RESUMO

PURPOSE: The mechanisms involved in the reported beneficial effects of Calcium dobesilate monohydrate (CaD) for the treatment of diabetic retinopathy (DR) remain to be elucidated. The main aim of the present study is to examine whether CaD prevents early events in the pathogenesis of DR such as neurodegeneration and vascular leakage. In addition, putative mediators of both neurodegeneration (glutamate/GLAST, ET-1/ETB receptor) and early microvascular impairment (ET-1/ETA receptor, oxidative stress, VEGF, and the PKC-delta-p38 MAPK pathway) have been examined. METHODS: Diabetic (db/db) mice were randomly assigned to daily oral treatment with CaD (200 mg/Kg/day) (n = 12) or vehicle (n = 12) for 14 days. In addition, 12 non-diabetic (db/+) mice matched by age were used as the control group. Functional abnormalities were assessed by electroretinography. Neurodegeneration and microvascular abnormalities were evaluated by immunohistochemistry and Western blot. Glutamate was determined by HPLC. RESULTS: CaD significantly decreased glial activation and apoptosis and produced a significant improvement in the electroretinogram parameters. Mechanistically, CaD prevented the diabetes-induced up-regulation of ET-1 and its cognate receptors (ETA-R and ETB-R), which are involved in microvascular impairment and neurodegeneration, respectively. In addition, treatment with CaD downregulated GLAST, the main glutamate transporter, and accordingly prevented the increase in glutamate. Finally, CaD prevented oxidative stress, and the upregulation of VEGF and PKC delta-p38 MAPK pathway induced by diabetes, thus resulting in a significant reduction in vascular leakage. CONCLUSIONS: Our findings demonstrate for the first time that CaD exerts neuroprotection in an experimental model of DR. In addition, we provide first evidence that CaD prevents the overexpression of ET-1 and its receptors in the diabetic retina. These beneficial effects on the neurovascular unit could pave the way for clinical trials addressed to confirm the effectiveness of CaD in very early stages of DR.


Assuntos
Dobesilato de Cálcio/farmacologia , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2/complicações , Retinopatia Diabética/prevenção & controle , Estresse Oxidativo/genética , Retina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Sobrevivência Celular , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Retinopatia Diabética/diagnóstico , Retinopatia Diabética/etiologia , Eletrorretinografia , Ensaio de Imunoadsorção Enzimática , Regulação da Expressão Gênica , Hemostáticos/farmacologia , Humanos , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Microscopia de Fluorescência , Estresse Oxidativo/efeitos dos fármacos , RNA/genética , Reação em Cadeia da Polimerase em Tempo Real , Retina/efeitos dos fármacos , Retina/patologia , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
11.
Front Med (Lausanne) ; 1: 41, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25593914

RESUMO

Secondary bacterial infections following influenza infection are a pressing problem facing respiratory medicine. Although antibiotic treatment has been highly successful over recent decades, fatalities due to secondary bacterial infections remain one of the leading causes of death associated with influenza. We have assessed whether administration of a bacterial extract alone is sufficient to potentiate immune responses and protect against primary infection with influenza, and secondary infections with either Streptococcus pneumoniae or Klebsiella pneumoniae in mice. We show that oral administration with the bacterial extract, OM-85, leads to a maturation of dendritic cells and B-cells characterized by increases in MHC II, CD86, and CD40, and a reduction in ICOSL. Improved immune responsiveness against influenza virus reduced the threshold of susceptibility to secondary bacterial infections, and thus protected the mice. The protection was associated with enhanced polyclonal B-cell activation and release of antibodies that were effective at neutralizing the virus. Taken together, these data show that oral administration of bacterial extracts provides sufficient mucosal immune stimulation to protect mice against a respiratory tract viral infection and associated sequelae.

12.
PLoS One ; 8(12): e82867, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24386121

RESUMO

OM-85 (Broncho-Vaxom®, Broncho-Munal®, Ommunal®, Paxoral®, Vaxoral®), a product made of the water soluble fractions of 21 inactivated bacterial strain patterns responsible for respiratory tract infections, is used for the prevention of recurrent upper respiratory tract infections and acute exacerbations in chronic obstructive pulmonary disease patients. OM-85 is able to potentiate both innate and adaptive immune responses. However, the molecular mechanisms responsible for OM-85 activation are still largely unknown. Purpose of this study was to investigate the impact of OM-85 stimulation on human dendritic cell functions. We show that OM-85 selectively induced NF-kB and MAPK activation in human DC with no detectable action on the interferon regulatory factor (IRF) pathway. As a consequence, chemokines (i.e. CXCL8, CXCL6, CCL3, CCL20, CCL22) and B-cell activating cytokines (i.e. IL-6, BAFF and IL-10) were strongly upregulated. OM-85 also synergized with the action of classical pro-inflammatory stimuli used at suboptimal concentrations. Peripheral blood mononuclear cells from patients with COPD, a pathological condition often associated with altered PRR expression pattern, fully retained the capability to respond to OM-85. These results provide new insights on the molecular mechanisms of OM-85 activation of the immune response and strengthen the rational for its use in clinical settings.


Assuntos
Extratos Celulares/farmacologia , Células Dendríticas/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases , NF-kappa B/fisiologia , Quimiocinas/genética , Quimiocinas/metabolismo , Células Dendríticas/fisiologia , Sinergismo Farmacológico , Humanos , Fatores Reguladores de Interferon/efeitos dos fármacos , Leucócitos Mononucleares/efeitos dos fármacos , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Doença Pulmonar Obstrutiva Crônica/sangue , Regulação para Cima
13.
J Neuroinflammation ; 9: 76, 2012 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-22533966

RESUMO

BACKGROUND: Cerebral ischemia is associated with the activation of glial cells, infiltration of leukocytes and an increase in inflammatory mediators in the ischemic brain and systemic circulation. How this inflammatory response influences lesion size and neurological outcome remains unclear. D-JNKI1, an inhibitor of the c-Jun N-terminal kinase pathway, is strongly neuroprotective in animal models of stroke. Intriguingly, the protection mediated by D-JNKI1 is high even with intravenous administration at very low doses with undetectable drug levels in the brain, pointing to a systemic mode of action, perhaps on inflammation. FINDINGS: We evaluated whether D-JNKI1, administered intravenously 3 h after the onset of middle cerebral artery occlusion (MCAO), modulates secretion of the inflammatory mediators interleukin-6 and keratinocyte-derived chemokine in the plasma and from the spleen and brain at several time points after MCAO. We found an early release of both mediators in the systemic circulation followed by an increase in the brain and went on to show a later systemic increase in vehicle-treated mice. Release of interleukin-6 and keratinocyte-derived chemokine from the spleen of mice with MCAO was not significantly different from sham mice. Interestingly, the secretion of these inflammatory mediators was not altered in the systemic circulation or brain after successful neuroprotection with D-JNKI1. CONCLUSIONS: We demonstrate that neuroprotection with D-JNKI1 after experimental cerebral ischemia is independent of systemic and brain release of interleukin-6 and keratinocyte-derived chemokine. Furthermore, our findings suggest that the early systemic release of interleukin-6 and keratinocyte-derived chemokine may not necessarily predict an unfavorable outcome in this model.


Assuntos
Isquemia Encefálica/prevenção & controle , Quimiocina CXCL1/metabolismo , Quimiocinas/metabolismo , Interleucina-6/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Fármacos Neuroprotetores/uso terapêutico , Peptídeos/uso terapêutico , Animais , Isquemia Encefálica/enzimologia , Isquemia Encefálica/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Valor Preditivo dos Testes , Distribuição Aleatória
14.
Eur J Immunol ; 39(4): 1136-46, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19291697

RESUMO

Phosphoinositide 3-kinase gamma (PI3Kgamma) plays a fundamental role in mediating leukocyte migration to inflammation sites. However, the downstream cytoplasmic events triggered by its signaling activity are still largely obscure. To address this issue, tyrosine and serine/threonine phosphorylated proteins of chemokine-stimulated WT or PI3Kgamma-null macrophages were investigated. Among the proteins analyzed, the intermediate filament vimentin was found as a downstream effector of the PI3Kgamma signaling pathway. Specific analysis of the phosphorylation state of vimentin in macrophages showed that this protein becomes rapidly phosphorylated in both tyrosine and serine residues upon chemokine stimulation. In the absence of PI3Kgamma or the kinase activity of PI3Kgamma (PI3Kgamma(KD/KD)), phosphorylation of vimentin was reduced. PI3Kgamma-null macrophages displayed impaired chemokine-driven vimentin fiber disassembly as well as reduced ability to transmigrate across endothelial cells. While WT macrophages infected with a vimentin mutant resistant to N-terminal serine phosphorylation showed a reduction in transendothelial migration, infection of PI3Kgamma-null macrophages with a vimentin mutant mimicking serine phosphorylation of N-terminal residues rescued the transendothelial migration defect. These results define vimentin N-terminal phosphorylation and fiber reorganization as a target of chemokine-dependent PI3Kgamma signaling in leukocytes.


Assuntos
Movimento Celular/imunologia , Leucócitos/imunologia , Macrófagos/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Vimentina/metabolismo , Animais , Classe Ib de Fosfatidilinositol 3-Quinase , Isoenzimas/genética , Isoenzimas/metabolismo , Leucócitos/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Camundongos Mutantes , Fosfatidilinositol 3-Quinases/genética , Fosforilação , Transdução de Sinais/imunologia , Vimentina/genética
15.
Proteomics ; 8(21): 4560-76, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18846510

RESUMO

Chemokines (CCs) are small chemoattractant cytokines involved in a wide variety of biological and pathological processes. Released by cells in the milieu, and extracellular matrix and activating signalling cascades upon binding to specific G protein-coupled receptors (GPCRs), they trigger many cellular events. In various pathologies, CCs are directly responsible for excessive recruitment of leukocytes to inflammatory sites and recent studies using chemokine receptor (CCR) antagonists permitted these molecules to reach the market for medical use. While interaction of CCs with their receptors has been extensively documented, downstream GPCR signalling cascades triggered by CC are less well understood. Given the pivotal role of chemokine receptor 2 (CCR2) in monocyte recruitment, activation and differentiation and its implication in several autoimmune-inflammatory pathologies, we searched for potential new CCR2-interacting proteins by engineering a modified CCR2 that we used as bait. Herein, we show the direct interaction of CCR2 with transportin1 (TRN1), which we demonstrate is followed by CCR2 receptor internalization. Further characterization of this novel interaction revealed that TRN1-binding to CCR2 increased upon time in agonist treated cells and promotes its nuclear translocation in a TRN1-dependent manner. Finally, we provide evidence that following translocation, the receptor localizes at the outer edge of the nuclear envelope where it is finally released from TRN1.


Assuntos
Núcleo Celular/metabolismo , Receptores CCR2/metabolismo , beta Carioferinas/fisiologia , Transporte Ativo do Núcleo Celular , Linhagem Celular , Quimiocina CCL2/metabolismo , Quimiotaxia , Cromatografia Líquida , Epitopos , Hemaglutininas/metabolismo , Humanos , Imunoprecipitação , Mapeamento de Interação de Proteínas , Proteômica , RNA Interferente Pequeno/genética , Transdução de Sinais , Espectrometria de Massas em Tandem , beta Carioferinas/agonistas
16.
J Immunol ; 180(4): 2538-44, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18250464

RESUMO

The leukocyte-enriched p110gamma and p110delta isoforms of PI3K have been shown to control in vitro degranulation of mast cells induced by cross-linking of the high affinity receptor of IgE (FcepsilonRI). However, the relative contribution of these PI3K isoforms in IgE-dependent allergic responses in vivo is controversial. A side-by-side comparative analysis of the role of p110gamma and p110delta in mast cell function, using genetic approaches and newly developed isoform-selective pharmacologic inhibitors, confirms that both PI3K isoforms play an important role in FcepsilonRI-activated mast cell degranulation in vitro. In vivo, however, only p110delta was found to be required for optimal IgE/Ag-dependent hypersensitivity responses in mice. These observations identify p110delta as a key therapeutic target among PI3K isoforms for allergy- and mast cell-related diseases.


Assuntos
Hipersensibilidade/enzimologia , Hipersensibilidade/imunologia , Fosfatidilinositol 3-Quinases/fisiologia , Animais , Domínio Catalítico/efeitos dos fármacos , Domínio Catalítico/genética , Degranulação Celular/efeitos dos fármacos , Degranulação Celular/imunologia , Células Cultivadas , Classe I de Fosfatidilinositol 3-Quinases , Epitopos/fisiologia , Hipersensibilidade/genética , Hipersensibilidade/patologia , Imunoglobulina E/fisiologia , Mediadores da Inflamação/administração & dosagem , Mediadores da Inflamação/farmacologia , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Isoenzimas/fisiologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Masculino , Mastócitos/efeitos dos fármacos , Mastócitos/enzimologia , Mastócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfatidilinositol 3-Quinases/genética , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/deficiência , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de IgG/fisiologia
17.
Mol Cell Proteomics ; 6(11): 1829-41, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17617665

RESUMO

Prior work using lipid-based affinity matrices has been done to investigate distinct sets of lipid-binding proteins, and one series of experiments has proven successful in mammalian cells for the proteome-wide identification of lipid-binding proteins. However, most lipid-based proteomics screens require scaled up sample preparation, are often composed of multiple cell types, and are not adapted for simultaneous signal transduction studies. Herein we provide a chemical proteomics strategy that uses cleavable lipid "baits" with broad applicability to diverse biological samples. The novel baits were designed to avoid preparative steps to allow functional proteomics studies when the biological source is a limiting factor. Validation of the chemical baits was first confirmed by the selective isolation of several known endogenous phosphatidylinositol 3-kinase signaling proteins using primary bone marrow-derived macrophages. The use of this technique for cellular proteomics and MS/MS analysis was then demonstrated by the identification of known and potential novel lipid-binding proteins that was confirmed in vitro for several proteins by direct lipid-protein interactions. Further to the identification, the method is also compatible with subsequent signal transduction studies, notably for protein kinase profiling of the isolated lipid-bound protein complexes. Taken together, this integration of minimal scale proteomics, lipid chemistry, and activity-based readouts provides a significant advancement in the ability to identify and study the lipid proteome of single, relevant cell types.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/isolamento & purificação , Macrófagos/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositóis/química , Proteômica/métodos , Animais , Células Cultivadas , Cromatografia de Afinidade , Peptídeos e Proteínas de Sinalização Intracelular/química , Lipídeos/química , Camundongos , Transdução de Sinais
18.
Eur J Immunol ; 35(5): 1501-9, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15789355

RESUMO

Treatment (from 5 to 25 weeks of age) with a novel blocking monoclonal antibody, mAb I-10, directed against the plasma membrane (pm) form of LAMP-1, protected against development of autoimmune diabetes in the NOD mouse. A shorter course of treatment, i.e. from 5 to 12 weeks of age, significantly reduced the occurrence of insulitis as well as disease onset. Interfering with pm-LAMP-1 required continuous treatment as tolerance was not observed when treatment was stopped, and no higher proportion of cells with a T regulatory phenotype (e.g. CD4(+)CD25(+)) were induced. The mechanism appears to involve modulating a proinflammatory cytokine, as the proportion of pancreatic-infiltrating IFN-gamma-positive cells was significantly reduced in the mAb I-10-treated group. These results demonstrate an unexpected role for pm-LAMP-1 in autoimmune disease progression, and suggest that further investigation should be performed to understand how this molecule modulates IFN-gamma-driven responses.


Assuntos
Anticorpos Bloqueadores/imunologia , Anticorpos Monoclonais/imunologia , Antígenos CD/imunologia , Diabetes Mellitus Tipo 1/imunologia , Proteínas de Membrana/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Bloqueadores/farmacologia , Anticorpos Monoclonais/farmacologia , Western Blotting , Diabetes Mellitus Tipo 1/prevenção & controle , Modelos Animais de Doenças , Eletroforese em Gel de Poliacrilamida , Feminino , Citometria de Fluxo , Interferon gama/biossíntese , Interferon gama/efeitos dos fármacos , Interferon gama/imunologia , Proteínas de Membrana Lisossomal , Proteínas de Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos NOD , Dados de Sequência Molecular , Pâncreas/efeitos dos fármacos , Pâncreas/imunologia , Células Th1/efeitos dos fármacos , Células Th1/imunologia
19.
J Biol Chem ; 279(41): 43273-84, 2004 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-15292195

RESUMO

In macrophages, chemotactic stimuli cause the activation of Rac and PAK, but little is known about the signaling pathways involved and their role in chemotactic gradient sensing. Herein, we report that in macrophages, the chemokine RANTES (regulated on activation normal T cell expressed and secreted)/CCL5 activates the small GTPase Rac and its downstream target PAK2 within seconds. This response depends on Gi activation and largely on the subsequent triggering of phosphoinositide 3-kinase gamma (PI3Kgamma) and Rac. Retroviral transduction of tagged Rac1 and -2 indicates that RANTES/CCL5-mediated activation of PI3Kgamma triggers Rac1 but not Rac2. In agreement, silencing of Rac1 by shRNA blocks PAK2 activity and inhibits RANTES/CCL5-induced macrophage polarization and directional migration. On the other hand, the tyrosine kinase receptor agonist CSF-1 activates PAK2 independently of PI3Kgamma and Rac. Our results thus demonstrate a chemokine-specific signaling pathway in which Gi and PI3Kgamma coordinate to drive Rac1 and PAK2 activation that eventually controls the chemotactic response.


Assuntos
Quimiocinas/metabolismo , Isoenzimas/fisiologia , Macrófagos/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Animais , Movimento Celular , Separação Celular , Quimiocina CCL5/metabolismo , Quimiotaxia , Cromonas/farmacologia , Classe Ib de Fosfatidilinositol 3-Quinase , Ativação Enzimática , Epitopos , Citometria de Fluxo , Inativação Gênica , Genisteína/farmacologia , Immunoblotting , Fator Estimulador de Colônias de Macrófagos/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia de Vídeo , Modelos Biológicos , Morfolinas/farmacologia , Toxina Pertussis/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Estrutura Terciária de Proteína , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt , RNA/química , RNA Mensageiro/metabolismo , Retroviridae/genética , Transdução de Sinais , Fatores de Tempo , Quinases Ativadas por p21
20.
Mol Endocrinol ; 17(11): 2228-39, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12907755

RESUMO

Protein tyrosine phosphatases (PTPs) play key roles in switching off tyrosine phosphorylation cascades, such as initiated by cytokine receptors. We have used substrate-trapping mutants of a large set of PTPs to identify members of the PTP family that have substrate specificity for the phosphorylated human GH receptor (GHR) intracellular domain. Among 31 PTPs tested, T cell (TC)-PTP, PTP-beta, PTP1B, stomach cancer-associated PTP 1 (SAP-1), Pyst-2, Meg-2, and PTP-H1 showed specificity for phosphorylated GHR that had been produced by coexpression with a kinase in bacteria. We then used GH-induced, phosphorylated GH receptor, purified from overexpressing mammalian cells, in a Far Western-based approach to test whether these seven PTPs were also capable of recognizing ligand-induced, physiologically phosphorylated GHR. In this assay, only TC-PTP, PTP1B, PTP-H1, and SAP-1 interacted with the mature form of the phosphorylated GHR. In parallel, we show that these PTPs recognize very different subsets of the seven GHR tyrosines that are potentially phosphorylated. Finally, mRNA tissue distribution of these PTPs by RT-PCR analysis and coexpression of the wild-type PTPs to test their ability to dephosphorylate ligand-activated GHR suggest PTP-H1 and PTP1B as potential candidates involved in GHR signaling.


Assuntos
Proteínas Tirosina Fosfatases/metabolismo , Receptores da Somatotropina/agonistas , Receptores da Somatotropina/metabolismo , Sequência de Aminoácidos , Animais , Células CHO , Linhagem Celular , Cricetinae , Hormônio do Crescimento/metabolismo , Humanos , Ligantes , Dados de Sequência Molecular , Especificidade de Órgãos , Fosfotirosina/metabolismo , Receptores da Somatotropina/química , Receptores da Somatotropina/genética , Transdução de Sinais , Especificidade por Substrato , Tirosina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...