Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Stem Cells Transl Med ; 13(4): 346-361, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38381583

RESUMO

BACKGROUND: In preclinical studies, mesenchymal stromal cells (MSCs), including umbilical cord-derived MSCs (UC-MSCs), demonstrate the ability to modulate numerous pathophysiological processes related to sepsis; however, a systematic synthesis of the literature is needed to assess the efficacy of UC-MSCs for treating sepsis. OBJECTIVE: To examine the effects of UC-MSCs on overall mortality (primary outcome) as well as on organ dysfunction, coagulopathy, endothelial permeability, pathogen clearance, and systemic inflammation (secondary outcomes) at prespecified time intervals in preclinical models of sepsis. METHODS: A systematic search was conducted on Embase, Ovid MEDLINE, and Web of Science up to June 20, 2023. Preclinical controlled studies using in vivo sepsis models with systemic UC-MSC administration were included. Meta-analyses were conducted and expressed as odds ratios (OR) and ratios of the weighted means with 95% CI for categorical and continuous data, respectively. Risk of bias was assessed with the SYRCLE tool. RESULTS: Twenty-six studies (34 experiments, n = 1258 animals) were included in this review. Overall mortality was significantly reduced with UC-MSC treatment as compared to controls (OR: 0.26, 95% CI: 0.18-0.36). At various prespecified time intervals, UC-MSCs reduced surrogate measures of organ dysfunction related to the kidney, liver, and lung; reduced coagulopathy and endothelial permeability; and enhanced pathogen clearance from multiple sites. UC-MSCs also modulated systemic inflammatory mediators. No studies were rated as low risk across all SYCLE domains. CONCLUSIONS: These results demonstrate the efficacy of UC-MSC treatment in preclinical sepsis models and highlight their potential as a therapeutic intervention for septic shock.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Sepse , Cordão Umbilical , Sepse/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Cordão Umbilical/citologia , Humanos , Animais , Células-Tronco Mesenquimais/citologia , Modelos Animais de Doenças
2.
Physiol Rep ; 10(9): e15286, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35510328

RESUMO

Acute respiratory distress syndrome (ARDS) is a lung injury characterized by noncardiogenic pulmonary edema and hypoxic respiratory failure. The purpose of this study was to investigate the effects of therapeutic hypothermia on short-term experimental ARDS. Twenty adult female Yorkshire pigs were divided into four groups (n = 5 each): normothermic control (C), normothermic injured (I), hypothermic control (HC), and hypothermic injured (HI). Acute respiratory distress syndrome was induced experimentally via intrapulmonary injection of oleic acid. Target core temperature was achieved in the HI group within 1 h of injury induction. Cardiorespiratory, histologic, cytokine, and metabolomic data were collected on all animals prior to and following injury/sham. All data were collected for approximately 12 h from the beginning of the study until euthanasia. Therapeutic hypothermia reduced injury in the HI compared to the I group (histological injury score = 0.51 ± 0.18 vs. 0.76 ± 0.06; p = 0.02) with no change in gas exchange. All groups expressed distinct phenotypes, with a reduction in pro-inflammatory metabolites, an increase in anti-inflammatory metabolites, and a reduction in inflammatory cytokines observed in the HI group compared to the I group. Changes to respiratory system mechanics in the injured groups were due to increases in lung elastance (E) and resistance (R) (ΔE from pre-injury = 46 ± 14 cmH2 O L-1 , p < 0.0001; ΔR from pre-injury: 3 ± 2 cmH2 O L-1  s- , p = 0.30) rather than changes to the chest wall (ΔE from pre-injury: 0.7 ± 1.6 cmH2 O L-1 , p = 0.99; ΔR from pre-injury: 0.6 ± 0.1 cmH2 O L-1  s- , p = 0.01). Both control groups had no change in respiratory mechanics. In conclusion, therapeutic hypothermia can reduce markers of injury and inflammation associated with experimentally induced short-term ARDS.


Assuntos
Hipotermia Induzida , Lesão Pulmonar , Síndrome do Desconforto Respiratório , Animais , Biomarcadores , Citocinas , Feminino , Pulmão/patologia , Síndrome do Desconforto Respiratório/terapia , Mecânica Respiratória , Suínos
3.
Sci Rep ; 12(1): 8294, 2022 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-35585165

RESUMO

Follicular lymphoma (FL) is a cancer of B-cells, representing the second most common type of non-Hodgkin lymphoma and typically diagnosed at advanced stage in older adults. In contrast to the wide range of available molecular genetic data, limited data relating the metabolomic features of follicular lymphoma are known. Metabolomics is a promising analytical approach employing metabolites (molecules < 1 kDa in size) as potential biomarkers in cancer research. In this pilot study, we performed proton nuclear magnetic resonance spectroscopy (1H-NMR) on 29 cases of FL and 11 control patient specimens. The resulting spectra were assessed by both unsupervised and supervised statistical methods. We report significantly discriminant metabolomic models of common metabolites distinguishing FL from control tissues. Within our FL case series, we also report discriminant metabolomic signatures predictive of progression-free survival.


Assuntos
Linfoma Folicular , Idoso , Humanos , Linfonodos , Linfoma Folicular/diagnóstico , Espectroscopia de Ressonância Magnética/métodos , Metabolômica/métodos , Projetos Piloto
4.
Neurocrit Care ; 35(3): 617-630, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34061312

RESUMO

BACKGROUND: Neurological injury can alter the systemic immune system, modifying the functional capacity of immune cells and causing a dysfunctional balance of cytokines, although mechanisms remain incompletely understood. The objective of this study was to assess the temporal relationship between changes in the activation status of circulating invariant natural killer T (iNKT) cells and the balance of plasma cytokines among critically ill patients with neurological injury. METHODS: We conducted an exploratory prospective observational study of adult (18 years or older) intensive care unit (ICU) patients with acute neurological injury (n = 20) compared with ICU patients without neurological injury (n = 22) and healthy controls (n = 10). Blood samples were collected on days 1, 2, 4, 7, 14, and 28 following ICU admission to analyze the activation status of circulating iNKT cells by flow cytometry and the plasma concentration of inflammation-relevant immune mediators, including T helper 1 (TH1) and T helper 2 (TH2) cytokines, by multiplex bead-based assay. RESULTS: Invariant natural killer T cells were activated in both ICU patient groups compared with healthy controls. Neurological patients had decreased levels of multiple immune mediators, including TH1 cytokines (interferon-γ, tumor necrosis factor-α, and interleukin-12p70), indicative of immunosuppression. This led to a greater than twofold increase in the ratio of TH2/TH1 cytokines early after injury (days 1 - 2) compared with healthy controls, a shift that was also observed for ICU controls. Systemic TH2/TH1 cytokine ratios were positively associated with iNKT cell activation in the neurological patients and negatively associated in ICU controls. These relationships were strongest for the CD4+ iNKT cell subset compared with the CD4- iNKT cell subset. The relationships to individual cytokines similarly differed between patient groups. Forty percent of the neurological patients developed an infection; however, differences for the infection subgroup were not identified. CONCLUSIONS: Critically ill patients with neurological injury demonstrated altered systemic immune profiles early after injury, with an association between activated peripheral iNKT cells and elevated systemic TH2/TH1 cytokine ratios. This work provides further support for a brain-immune axis and the ability of neurological injury to have far-reaching effects on the body's immune system.


Assuntos
Células T Matadoras Naturais , Estado Terminal , Citocinas , Citometria de Fluxo , Humanos , Interferon gama
5.
Sci Rep ; 9(1): 19584, 2019 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-31863066

RESUMO

Sarcoidosis is a disorder characterized by granulomatous inflammation of unclear etiology. In this study we evaluated whether veterans with sarcoidosis exhibited different plasma metabolomic and metallomic profiles compared with civilians with sarcoidosis. A case control study was performed on veteran and civilian patients with confirmed sarcoidosis. Proton nuclear magnetic resonance spectroscopy (1H NMR), hydrophilic interaction liquid chromatography mass spectrometry (HILIC-MS) and inductively coupled plasma mass spectrometry (ICP-MS) were applied to quantify metabolites and metal elements in plasma samples. Our results revealed that the veterans with sarcoidosis significantly differed from civilians, according to metabolic and metallomics profiles. Moreover, the results showed that veterans with sarcoidosis and veterans with COPD were similar to each other in metabolomics and metallomics profiles. This study suggests the important role of environmental risk factors in the development of different molecular phenotypic responses of sarcoidosis. In addition, this study suggests that sarcoidosis in veterans may be an occupational disease.


Assuntos
Metabolômica , Metais/química , Doença Pulmonar Obstrutiva Crônica/metabolismo , Sarcoidose Pulmonar/metabolismo , Estudos de Casos e Controles , Cromatografia Líquida , Análise Discriminante , Feminino , Humanos , Análise dos Mínimos Quadrados , Espectroscopia de Ressonância Magnética , Masculino , Espectrometria de Massas , Fenótipo , Prótons , Doença Pulmonar Obstrutiva Crônica/diagnóstico , Estudos Retrospectivos , Sarcoidose Pulmonar/diagnóstico , Pesquisa Translacional Biomédica , Estados Unidos , Veteranos
6.
Crit Care Med ; 47(7): 918-925, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30720538

RESUMO

OBJECTIVES: Cellular Immunotherapy for Septic Shock is the first-in-human clinical trial evaluating allogeneic mesenchymal stem/stromal cells in septic shock patients. Here, we sought to determine whether plasma cytokine profiles may provide further information into the safety and biological effects of mesenchymal stem/stromal cell treatment, as no previous study has conducted a comprehensive analysis of circulating cytokine levels in critically ill patients treated with mesenchymal stem/stromal cells. DESIGN: Phase 1 dose-escalation trial. PATIENTS: The interventional cohort (n = 9) of septic shock patients received a single dose of 0.3, 1.0, or 3.0 million mesenchymal stem/stromal cells/kg body weight (n = 3 per dose). The observational cohort received no mesenchymal stem/stromal cells (n = 21). INTERVENTIONS: Allogeneic bone marrow-derived mesenchymal stem/stromal cells. MEASUREMENTS AND MAIN RESULTS: Serial plasma samples were collected at study baseline prior to mesenchymal stem/stromal cell infusion (0 hr), 1 hour, 4 hours, 12 hours, 24 hours, and 72 hours after mesenchymal stem/stromal cell infusion/trial enrollment. Forty-nine analytes comprised mostly of cytokines along with several biomarkers were measured. We detected no significant elevations in a broad range of pro-inflammatory cytokines and biomarkers between the interventional and observational cohorts. Stratification of the interventional cohort by mesenchymal stem/stromal cell dose further revealed patient-specific and dose-dependent perturbations in cytokines, including an early but transient dampening of pro-inflammatory cytokines (e.g., interleukin-1ß, interleukin-2, interleukin-6, interleukin-8, and monocyte chemoattractant protein 1), suggesting that mesenchymal stem/stromal cell treatment may alter innate immune responses and underlying sepsis biology. CONCLUSIONS: A single infusion of up to 3 million cells/kg of allogeneic mesenchymal stem/stromal cells did not exacerbate elevated cytokine levels in plasma of septic shock patients, consistent with a safe response. These data also offer insight into potential biological mechanisms of mesenchymal stem/stromal cell treatment and support further investigation in larger randomized controlled trials.


Assuntos
Citocinas/biossíntese , Transplante de Células-Tronco Mesenquimais/métodos , Choque Séptico/metabolismo , Choque Séptico/terapia , Adulto , Biomarcadores , Relação Dose-Resposta a Droga , Feminino , Humanos , Mediadores da Inflamação/metabolismo , Masculino , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Pessoa de Meia-Idade , Índice de Gravidade de Doença
7.
Neurocrit Care ; 30(1): 22-32, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-29569129

RESUMO

This scoping review will discuss the basic functions and prognostic significance of the commonly researched cytokines implicated in severe traumatic brain injury (sTBI), including tumour necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), IL-6, tissue inhibitor of matrix metalloproteinases-1 (TIMP-1), transforming growth factor-ß (TGF-ß), substance P, and soluble CD40 ligand (sCD40L). A scoping review was undertaken with an electronic search for articles from the Ovid MEDLINE, PUBMED and EMBASE databases from 1995 to 2017. Inclusion criteria were original research articles, and reviews including both animal models and human clinical studies of acute (< 3 months) sTBI. Selected articles included both isolated sTBI and sTBI with systemic injury. After applying the inclusion criteria and removing duplicates, 141 full-text articles, 126 original research articles and 15 review articles, were evaluated in compiling this review paper. A single reviewer, CC, completed the review in two phases. During the first phase, titles and abstracts of selected articles were reviewed for inclusion. A second evaluation was then conducted on the full text of all selected articles to ensure relevancy. From our current understanding of the literature, it is unlikely a single biomarker will be sufficient in accurately prognosticating patients with sTBI. Intuitively, a more severe injury will demonstrate higher levels of inflammatory cytokines which may correlate as a marker of severe injury. This does not mean, necessarily, these cytokines have a direct and causal role in the poor outcome of the patient. Further research is required to better delineate the complex systemic inflammatory and CNS interactions that occur during sTBI before they can be applied as a reliable prognostic tool.


Assuntos
Biomarcadores/metabolismo , Lesões Encefálicas Traumáticas/diagnóstico , Citocinas/metabolismo , Animais , Lesões Encefálicas Traumáticas/imunologia , Lesões Encefálicas Traumáticas/metabolismo , Humanos
8.
Am J Respir Crit Care Med ; 197(3): 337-347, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28960096

RESUMO

RATIONALE: In septic animal models mesenchymal stem (stromal) cells (MSCs) modulate inflammation, enhance tissue repair and pathogen clearance, and reduce death. OBJECTIVES: To conduct a phase I dose escalation trial of MSCs in septic shock with the primary objective of examining the safety and tolerability of MSCs. METHODS: We enrolled nine participants within 24 hours of admission to the ICU. A control cohort of 21 participants was enrolled before starting the MSC interventional cohort to characterize expected adverse events (AEs) and to serve as a comparator for the intervention cohort. Three separate MSC dose cohorts, with three participants per cohort, received a single intravenous dose of 0.3, 1.0, and 3.0 × 106 cells/kg. A prespecified safety plan monitored participants for the occurrence of AEs; cytokines were collected at prespecified time points. MEASUREMENTS AND MAIN RESULTS: Ages of participants in the interventional versus observational cohorts were median of 71 (range, 38-91) and 61 (range, 23-95). Acute Physiology and Chronic Health Evaluation scores were median of 25 (range, 11-28) and 26 (range, 17-32). MSC doses ranged from 19 to 250 million cells. There were no prespecified MSC infusion-associated or serious unexpected AEs, nor any safety or efficacy signals for the expected AEs or the measured cytokines between the interventional and observational cohorts. CONCLUSIONS: The infusion of freshly cultured allogenic bone marrow-derived MSCs, up to a dose of 3 million cells/kg (250 million cells), into participants with septic shock seems safe. Clinical trial registered with www.clinicaltrials.gov (NCT02421484).


Assuntos
Imunoterapia/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Choque Séptico/terapia , Adulto , Fatores Etários , Idoso , Aloenxertos , Intervalos de Confiança , Feminino , Seguimentos , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Medição de Risco , Fatores Sexuais , Choque Séptico/diagnóstico , Choque Séptico/mortalidade , Taxa de Sobrevida , Resultado do Tratamento , Adulto Jovem
9.
PLoS One ; 7(10): e47559, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23133515

RESUMO

BACKGROUND: Mesenchymal stromal cells (MSCs, "adult stem cells") have been widely used experimentally in a variety of clinical contexts. There is interest in using these cells in critical illness, however, the safety profile of these cells is not well known. We thus conducted a systematic review of clinical trials that examined the use MSCs to evaluate their safety. METHODS AND FINDINGS: MEDLINE, EMBASE, and the Cochrane Central Register of Controlled Trials (to June 2011), were searched. Prospective clinical trials that used intravascular delivery of MSCs (intravenously or intra-arterially) in adult populations or mixed adult and pediatric populations were identified. Studies using differentiated MSCs or additional cell types were excluded. The primary outcome adverse events were grouped according to immediate events (acute infusional toxicity, fever), organ system complications, infection, and longer term adverse events (death, malignancy). 2347 citations were reviewed and 36 studies met inclusion criteria. A total of 1012 participants with clinical conditions of ischemic stroke, Crohn's disease, cardiomyopathy, myocardial infarction, graft versus host disease, and healthy volunteers were included. Eight studies were randomized control trials (RCTs) and enrolled 321 participants. Meta-analysis of the RCTs did not detect an association between acute infusional toxicity, organ system complications, infection, death or malignancy. There was a significant association between MSCs and transient fever. CONCLUSIONS: Based on the current clinical trials, MSC therapy appears safe. However, further larger scale controlled clinical trials with rigorous reporting of adverse events are required to further define the safety profile of MSCs.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Ensaios Clínicos como Assunto , Células-Tronco Mesenquimais/citologia , Células Estromais/citologia , Adolescente , Adulto , Idoso , Cardiomiopatias/terapia , Doença Enxerto-Hospedeiro/terapia , Humanos , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/terapia , Isquemia Miocárdica/terapia , Segurança do Paciente , Estudos Prospectivos , Ensaios Clínicos Controlados Aleatórios como Assunto , Resultado do Tratamento
10.
Am J Physiol Lung Cell Mol Physiol ; 303(9): L788-98, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22923639

RESUMO

Pulmonary fibrosis, the end stage of a variety of fibroproliferative lung diseases, is usually induced after repetitive or chronic lung injury or inflammation. The mechanisms of fibroproliferation are poorly understood. Insulin-like growth factor-I (IGF-I) is significantly elevated in patients with pulmonary fibrosis and fibroproliferative acute respiratory distress syndrome. However, we showed that IGF-I overexpression alone in wild-type mouse lungs does not cause fibroproliferation. We therefore questioned whether IGF-I, acting together with active TGF-ß1, a known profibrotic cytokine, enhances pulmonary fibroproliferation caused by active TGF-ß1. A unique sequential adenoviral transgene mouse model was used expressing AdEmpty/AdTGF-ß1 or AdhIGF-IB/AdTGF-ß1 transgenes. IGF-IB plus active TGF-ß1 transgene expression synergistically increased collagen deposition in the lung parenchyma compared with active TGF-ß1 expression alone. The enhanced fibrosis was accompanied by an increased recruitment of macrophages and lymphocytes into the bronchoalveolar lavage fluid (BALF) and inflammatory cells in the lungs. α-Smooth muscle actin expression, a marker of myofibroblast proliferation and differentiation, was also increased. Finally, fibroblasts exposed ex vivo to BALF isolated from AdhIGF-IB/AdTGF-ß1-transduced mice showed synergistic collagen induction compared with BALF from AdEmpty/AdTGF-ß1-transduced mice. This study provides the first direct evidence that IGF-I is able to synergistically enhance pulmonary fibroproliferation in cooperation with TGF-ß1.


Assuntos
Proliferação de Células , Fibroblastos/fisiologia , Fator de Crescimento Insulin-Like I/biossíntese , Pulmão/metabolismo , Fibrose Pulmonar/metabolismo , Fator de Crescimento Transformador beta1/biossíntese , Actinas/metabolismo , Animais , Líquido da Lavagem Broncoalveolar , Linhagem Celular , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Colágeno Tipo III/metabolismo , Citocinas/metabolismo , Fibroblastos/metabolismo , Expressão Gênica , Humanos , Mediadores da Inflamação/metabolismo , Fator de Crescimento Insulin-Like I/genética , Pulmão/imunologia , Pulmão/patologia , Linfócitos/imunologia , Linfócitos/patologia , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neutrófilos/imunologia , Neutrófilos/patologia , Fibrose Pulmonar/imunologia , Fibrose Pulmonar/patologia , Sus scrofa , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
11.
Am J Physiol Lung Cell Mol Physiol ; 298(4): L492-500, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20081067

RESUMO

Pulmonary fibrosis (PF), the end stage of a variety of fibroproliferative lung diseases, is characterized by excessive lung mesenchymal cell activation and extracellular matrix deposition. Most PF is induced after repetitive or chronic lung inflammation; however, a significant portion of PF occurs without apparent inflammation. The mechanisms of fibroproliferation are poorly understood. Studies have shown that cytokines regulating inflammation and tissue repair processes play essential roles in the development of PF. Insulin-like growth factor I (IGF-I) has been shown to stimulate lung mesenchymal cell proliferation and extracellular matrix synthesis in vitro and is significantly elevated in patients with PF. In this study, we investigated whether human IGF-IB (hIGF-IB) expression in the lungs induces PF in a C57BL/6 mouse model. Mice were subjected to adenoviral gene transfer, and the effects of hIGF-IB expression on the lungs were examined 3, 7, 14, 21, and 42 days after gene delivery. hIGF-IB expression induced significant and prolonged inflammatory cell infiltration into the lungs, with an early neutrophil infiltration followed by a late macrophage infiltration. No significant fibroblast or matrix accumulation could be detected in the lungs of these mice. No significant collagen accumulation could be detected in vivo, despite in vitro evidence that hIGF-IB induces collagen mRNA expression in fibroblasts. Therefore, IGF-IB alone is not sufficient to induce fibrosis, and it is possible that a coactivator is required to induce significant fibroproliferation in vivo.


Assuntos
Adenoviridae/genética , Inflamação/patologia , Fator de Crescimento Insulin-Like I/genética , Pulmão/metabolismo , Pulmão/patologia , Fibrose Pulmonar/patologia , Transdução Genética , Animais , Líquido da Lavagem Broncoalveolar/citologia , Movimento Celular , Proliferação de Células , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Colágeno Tipo III/genética , Colágeno Tipo III/metabolismo , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Humanos , Inflamação/complicações , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Isoformas de Proteínas/genética , Fibrose Pulmonar/complicações , Transcrição Gênica , Transgenes/genética
12.
Am J Physiol Cell Physiol ; 296(5): C1140-50, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19279234

RESUMO

To investigate the potential role of the local expression of alternative complement factor B (hBf) in human sepsis, we examined the induction of Bf gene expression in human peripheral blood monocytes (PBMCs) from patients with septic shock and the mechanisms of hBf gene regulation by tumor necrosis factor (TNF)-alpha, interferon (IFN)-gamma, and lipopolysaccharide (LPS) in human monocytes. PBMCs from septic shock patients showed increased hBf mRNA expression when compared with control patients. Costimulation with TNF-alpha and IFN-gamma or stimulation with LPS demonstrated a time- and dose-dependent induction of hBf mRNA expression in human PBMCs. A region of the hBf promoter between -735 and +128 bp was found to mediate IFN-gamma, TNF-alpha, and LPS responsiveness as well as the synergistic effect of IFN-gamma/TNF-alpha on hBf promoter activity. Site-directed mutagenesis of a IFN-gamma-activation site (GAS) cis element (-90 to -82 bp) abrogated IFN-gamma responsiveness. Mutagenesis of a nuclear factor (NF)-kappaB cis element at -466 to -456 bp abrogated TNF-alpha and LPS responsiveness of the Bf promoter. Thus hBf gene expression is induced in PBMCs from septic shock patients, and the induction of hBf by IFN-gamma, TNF-alpha, and LPS is through GAS and NF-kappaB cis-binding sites on the hBf promoter. Furthermore, activated protein C (APC) inhibited LPS-stimulated hBf promoter activity and protein expression in human monocytes suggesting that the beneficial effect of APC therapy in sepsis may in part be due to inhibition of complement induction and/or activation via the alternative pathway.


Assuntos
Fator B do Complemento/genética , Macrófagos/imunologia , Monócitos/imunologia , Proteína C/metabolismo , Sepse/imunologia , Sepse/fisiopatologia , Sequência de Bases , Fator B do Complemento/imunologia , Fator B do Complemento/metabolismo , Via Alternativa do Complemento/imunologia , Humanos , Interferon gama/imunologia , Interferon gama/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , Dados de Sequência Molecular , Monócitos/metabolismo , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/imunologia , Proteína C/imunologia , RNA Mensageiro/metabolismo , Sepse/metabolismo , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/imunologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
13.
J Virol ; 79(23): 14507-15, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16282450

RESUMO

In gene therapy, the innate immune system is a significant barrier to the effective application of adenovirus (Ad) vectors. In kidney epithelium-derived (REC) cells, serotype 5 Ad vectors induce the expression of the chemokine CXCL10 (IP-10), a response that is dependent on NFkappaB. Compared to the parental vector AdLuc, transduction with the RGD-deleted vector AdL.PB resulted in reduced CXCL10 activation despite increasing titers, implying that RGD-alpha(V) integrin interactions contribute to adenovirus induction of inflammatory genes. Akt, a downstream effector of integrin signaling, was activated within 10 min of transduction with Ad vectors in a dose-dependent manner. Akt activation was not present following transduction with AdL.PB, confirming the importance of capsid-alpha(V) integrin interactions in Ad vector Akt activation. Inhibition of the phosphoinositide-3-OH kinase/Akt pathway by Wortmannin or Ly294002 compounds decreased Ad vector induction of CXCL10 mRNA. Similarly, adenovirus-mediated overexpression of the dominant negative AktAAA decreased CXCL10 mRNA expression compared to the reporter vector AdLacZ alone. The effect of Akt on CXCL10 mRNA expression occurred via NFkappaB-dependent transcriptional activation, since AktAAA overexpression and Ly294002 both inhibited CXCL10 and NFkappaB promoter activation in luciferase reporter experiments. These results show that Akt plays a role in the Ad vector activation of NFkappaB and CXCL10 expression. Understanding the mechanism underlying the regulation of host immunomodulatory genes by adenovirus vectors will lead to strategies that will improve the efficacy and safety of these agents for clinical use.


Assuntos
Adenoviridae/fisiologia , Quimiocinas CXC/metabolismo , Regulação da Expressão Gênica , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Adenoviridae/genética , Adenoviridae/imunologia , Quimiocina CXCL10 , Quimiocinas CXC/genética , Vetores Genéticos , NF-kappa B/imunologia , Células Tumorais Cultivadas
14.
J Neurosci ; 24(6): 1521-9, 2004 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-14960625

RESUMO

The neuromodulator adenosine regulates immune activation and neuronal survival through specific G-protein-coupled receptors expressed on macrophages and neurons, including the A1 adenosine receptor (A1AR). Here we show that A1AR null (A1AR-/-) mice developed a severe progressive-relapsing form of experimental allergic encephalomyelitis (EAE) compared with their wild-type (A1AR+/+) littermates. Worsened demyelination, axonal injury, and enhanced activation of microglia/macrophages were observed in A1AR-/- animals. In addition, spinal cords from A1AR-/- mice demonstrated increased proinflammatory gene expression during EAE, whereas anti-inflammatory genes were suppressed compared with A1AR+/+ animals. Macrophages from A1AR-/- animals exhibited increased expression of the proinflammatory genes, interleukin-1beta, and matrix metalloproteinase-12 on immune activation when matched with A1AR+/+ control cells. A1AR-/- macrophage-derived soluble factors caused significant oligodendrocyte cytotoxicity compared with wild-type controls. The A1AR was downregulated in microglia in A1AR+/+ mice during EAE accompanied by neuroinflammation, which recapitulated findings in multiple sclerosis (MS) patients. Caffeine treatment augmented A1AR expression on microglia, with ensuing reduction of EAE severity, which was further enhanced by concomitant treatment with the A1AR agonist, adenosine amine congener. Thus, modulation of neuroinflammation by the A1AR represents a novel mechanism that provides new therapeutic opportunities for MS and other demyelinating diseases.


Assuntos
Adenosina/análogos & derivados , Encefalomielite Autoimune Experimental/fisiopatologia , Inflamação/metabolismo , Esclerose Múltipla/fisiopatologia , Bainha de Mielina/metabolismo , Receptor A1 de Adenosina/metabolismo , Regulação para Cima/fisiologia , Adenosina/farmacologia , Agonistas do Receptor A1 de Adenosina , Animais , Cafeína/farmacologia , Modelos Animais de Doenças , Progressão da Doença , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/patologia , Feminino , Humanos , Inflamação/imunologia , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Interleucina-1/genética , Interleucina-1/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Metaloproteinase 12 da Matriz , Metaloendopeptidases/genética , Metaloendopeptidases/metabolismo , Camundongos , Camundongos Knockout , Microglia/imunologia , Microglia/metabolismo , Microglia/patologia , Esclerose Múltipla/patologia , Bainha de Mielina/imunologia , Bainha de Mielina/patologia , Fenótipo , RNA Mensageiro/metabolismo , Receptor A1 de Adenosina/genética , Recidiva , Índice de Gravidade de Doença , Medula Espinal/patologia
15.
Am J Respir Crit Care Med ; 167(1): 83-90, 2003 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12502480

RESUMO

Insulin-like growth factor-I (IGF-I) is elevated in human fibrotic lung diseases and in animal models of pulmonary fibrosis, implicating IGF-I in the pathogenesis of fibrotic lung disease. We questioned whether IGF-I protein levels were enhanced in fibroproliferative acute respiratory distress syndrome (FP-ARDS). Serial lung tissue sections from a biopsy database were immunohistochemically stained for IGF-I, IGF-I receptor, CD68, alpha-smooth muscle actin, collagens I and III, and proliferating cell nuclear antigen. Our results show enhanced staining of IGF-I and IGF-I receptor, collagens I and III, smooth muscle actin, CD68, and proliferating cell nuclear antigen in FP-ARDS compared with control lung sections. In FP-ARDS specimens, prominent staining of IGF-I and IGF-I receptor was seen in alveolar and interstitial macrophages as well as in a variety of mesenchymal cells. There was a correlation between IGF-I staining and CD68-positive cells, suggesting macrophages as a potential source of the IGF-I protein present in lungs. IGF-I also correlated with enhanced collagen I, collagen III, and proliferating cell nuclear antigen immunoreactivity, suggesting that IGF-I may play a role in the extracellular matrix protein deposition and cellular proliferation seen in the lungs of individuals with FP-ARDS. Our results indicate that IGF-I is increased in FP-ARDS and may be an important mediator in the progression of acute lung injury to FP-ARDS.


Assuntos
Fator de Crescimento Insulin-Like I/análise , Pulmão/química , Síndrome do Desconforto Respiratório/metabolismo , Actinas/análise , Antígenos CD/análise , Antígenos de Diferenciação Mielomonocítica/análise , Colágeno Tipo I/análise , Colágeno Tipo III/análise , Feminino , Humanos , Imuno-Histoquímica , Macrófagos Alveolares/química , Masculino , Pessoa de Meia-Idade , Carbonitrila de Pregnenolona/análise , Receptor IGF Tipo 1/análise
16.
Chest ; 122(6 Suppl): 289S-293S, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12475802

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a lung disease that is characterized by epithelial cell damage and areas of denuded basement membrane resulting in inflammation, fibroblast proliferation, excessive extracellular matrix (ECM) deposition, and remodeling of alveolar gas exchange units. The progressive loss of lung gas exchange units in patients with IPF leads to respiratory failure and eventually to death. While the etiology of this disease is unknown, for many years studies suggested that chronic inflammation was the underlying factor that caused fibroproliferation and structural alterations of the lung. Recent data show that fibroproliferation and fibrosis can occur independently of inflammation, suggesting that IPF is a disease caused by a mesenchymal, rather than an immune disorder. Mesenchymal growth factors, including transforming growth factor (TGF)-beta, insulin-like growth factor (IGF)-I, platelet-derived growth factor, connective tissue growth factor, fibroblast growth factors, and keratinocyte growth factors, as well as proinflammatory cytokines such as tumor necrosis factor-alpha and interleukin-1beta, have been shown to be exaggerated in several fibrotic lung disorders including IPF, ARDS, sarcoidosis, and bronchopulmonary dysplasia, as well as pulmonary manifestations of systemic diseases such as rheumatoid arthritis or progressive systemic sclerosis (scleroderma). We argue that inflammation is required to initiate growth factor production and repair of the damaged alveolar epithelial lining in fibrotic lung diseases and that exaggerated TGF-beta production may be responsible for the fibrotic response seen in diseases such as IPF. We recognize the potential role of several growth factors in the fibroproliferative process in the lung, and in this brief report we focus on the possible roles of the growth factors IGF-I and TGF-beta in cell migration, proliferation, and ECM synthesis in patients with IPF.


Assuntos
Fator de Crescimento Insulin-Like I/fisiologia , Fibrose Pulmonar/fisiopatologia , Mucosa Respiratória/fisiopatologia , Fator de Crescimento Transformador beta/fisiologia , Humanos , Inflamação/fisiopatologia , Fibrose Pulmonar/imunologia , Fibrose Pulmonar/patologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/patologia
17.
J Immunol ; 169(5): 2627-35, 2002 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12193734

RESUMO

Complement factor B (Bf) plays an important role in activating the alternative complement pathway. The inflammatory cytokines, in particular TNF-alpha and IFN-gamma, are critical in the regulation of Bf gene expression in macrophages. In this study, we investigated the mechanisms of Bf gene regulation by TNF-alpha and IFN-gamma in murine macrophages. Northern analysis revealed that Bf mRNA expression was synergistically up-regulated by TNF-alpha and IFN-gamma in MH-S cells. Truncations of the 5' Bf promoter identified a region between -556 and -282 bp that mediated TNF-alpha responsiveness as well as the synergistic effect of TNF-alpha and IFN-gamma on Bf expression. Site-directed mutagenesis of a NF-kappaB-binding element in this region (-433 to -423 bp) abrogated TNF-alpha responsiveness and decreased the synergistic effect of TNF-alpha and IFN-gamma on Bf expression. EMSAs revealed nuclear protein binding to this NF-kappaB cis-binding element on TNF-alpha stimulation. Supershift analysis revealed that both p50 and p65 proteins contribute to induction of Bf by TNF-alpha. An I-kappaB dominant negative mutant blocked Bf induction by TNF-alpha and reduced the synergistic induction by TNF-alpha and IFN-gamma. In addition, the proteasome inhibitor MG132, which blocks NF-kappaB induction, blocked TNF-alpha-induced Bf promoter activity and the synergistic induction of Bf promoter activity by TNF-alpha and IFN-gamma. LPS was found to induce Bf promoter activity through the same NF-kappaB cis-binding site. These findings suggest that a NF-kappaB cis-binding site between -433 and -423 bp is required for TNF-alpha responsiveness and for TNF-alpha- and IFN-gamma-stimulated synergistic responsiveness of the Bf gene.


Assuntos
Fator B do Complemento/genética , Fator B do Complemento/metabolismo , Regulação da Expressão Gênica/imunologia , Interferon gama/fisiologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Regiões 5' não Traduzidas/fisiologia , Animais , Linhagem Celular , Núcleo Celular/genética , Núcleo Celular/imunologia , Núcleo Celular/metabolismo , Fator B do Complemento/biossíntese , Sinergismo Farmacológico , Proteínas I-kappa B/metabolismo , Proteínas I-kappa B/fisiologia , Interleucina-1/farmacologia , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos/genética , Camundongos , Mutagênese Sítio-Dirigida , NF-kappa B/biossíntese , NF-kappa B/genética , NF-kappa B/metabolismo , NF-kappa B/fisiologia , Subunidade p50 de NF-kappa B , Fosforilação , Regiões Promotoras Genéticas/imunologia , Ligação Proteica/genética , Ligação Proteica/imunologia , RNA Mensageiro/biossíntese , Elementos de Resposta/imunologia , Fator de Transcrição RelA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA