Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Lung Cell Mol Physiol ; 324(6): L783-L798, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37039367

RESUMO

NR2F2 is expressed in endothelial cells (ECs) and Nr2f2 knockout produces lethal cardiovascular defects. In humans, reduced NR2F2 expression is associated with cardiovascular diseases including congenital heart disease and atherosclerosis. Here, NR2F2 silencing in human primary ECs led to inflammation, endothelial-to-mesenchymal transition (EndMT), proliferation, hypermigration, apoptosis-resistance, and increased production of reactive oxygen species. These changes were associated with STAT and AKT activation along with increased production of DKK1. Co-silencing DKK1 and NR2F2 prevented NR2F2-loss-induced STAT and AKT activation and reversed EndMT. Serum DKK1 concentrations were elevated in patients with pulmonary arterial hypertension (PAH) and DKK1 was secreted by ECs in response to in vitro loss of either BMPR2 or CAV1, which are genetic defects associated with the development of PAH. In human primary ECs, NR2F2 suppressed DKK1, whereas its loss conversely induced DKK1 and disrupted endothelial homeostasis, promoting phenotypic abnormalities associated with pathologic vascular remodeling. Activating NR2F2 or blocking DKK1 may be useful therapeutic targets for treating chronic vascular diseases associated with EC dysfunction.NEW & NOTEWORTHY NR2F2 loss in the endothelial lining of blood vessels is associated with cardiovascular disease. Here, NR2F2-silenced human endothelial cells were inflammatory, proliferative, hypermigratory, and apoptosis-resistant with increased oxidant stress and endothelial-to-mesenchymal transition. DKK1 was induced in NR2F2-silenced endothelial cells, while co-silencing NR2F2 and DKK1 prevented NR2F2-loss-associated abnormalities in endothelial signaling and phenotype. Activating NR2F2 or blocking DKK1 may be useful therapeutic targets for treating vascular diseases associated with endothelial dysfunction.


Assuntos
Hipertensão Arterial Pulmonar , Doenças Vasculares , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células Endoteliais/metabolismo , Doenças Vasculares/metabolismo , Hipertensão Arterial Pulmonar/metabolismo , Hipertensão Pulmonar Primária Familiar/metabolismo , Inflamação/patologia , Fator II de Transcrição COUP/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo
2.
Monaldi Arch Chest Dis ; 92(1)2021 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-34461700

RESUMO

A pandemic caused by the Severe Acute Respiratory Syndrome Coronavirus 2 was declared in 2020. Severe cases were characterized by the development of acute hypoxemic respiratory failure (AHRF) requiring advanced respiratory support. However, intensive care units (ICU) were saturated, and many patients had to be treated out of ICU. This case describes a 75-year-old man affected by AHRF due to Coronavirus Disease 2019 (COVID-19), hospitalized in a high-dependency unit, with PaO2/FiO2 <100 for 28 consecutive days. An experienced team with respiratory physiotherapists was in charge of the noninvasive ventilatory support (NIVS). The patient required permanent NIVS with continuous positive airway pressure, non-invasive ventilation, high flow nasal oxygen and body positioning. He was weaned from NIVS after 37 days and started exercise training afterwards. The patient was discharged at home with low-flow oxygen therapy. This case represents an example of a successful treatment of AHRF with the still controversial noninvasive respiratory support in one patient with COVID-19.


Assuntos
COVID-19 , Ventilação não Invasiva , Insuficiência Respiratória , Idoso , Humanos , Masculino , Oxigenoterapia , Pandemias , Insuficiência Respiratória/etiologia , Insuficiência Respiratória/terapia
3.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-33836561

RESUMO

Interferonopathies, interferon (IFN)-α/ß therapy, and caveolin-1 (CAV1) loss-of-function have all been associated with pulmonary arterial hypertension (PAH). Here, CAV1-silenced primary human pulmonary artery endothelial cells (PAECs) were proliferative and hypermigratory, with reduced cytoskeletal stress fibers. Signal transducers and activators of transcription (STAT) and phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) were both constitutively activated in these cells, resulting in a type I IFN-biased inflammatory signature. Cav1-/- mice that spontaneously develop pulmonary hypertension were found to have STAT1 and AKT activation in lung homogenates and increased circulating levels of CXCL10, a hallmark of IFN-mediated inflammation. PAH patients with CAV1 mutations also had elevated serum CXCL10 levels and their fibroblasts mirrored phenotypic and molecular features of CAV1-deficient PAECs. Moreover, immunofluorescence staining revealed endothelial CAV1 loss and STAT1 activation in the pulmonary arterioles of patients with idiopathic PAH, suggesting that this paradigm might not be limited to rare CAV1 frameshift mutations. While blocking JAK/STAT or AKT rescued aspects of CAV1 loss, only AKT inhibitors suppressed activation of both signaling pathways simultaneously. Silencing endothelial nitric oxide synthase (NOS3) prevented STAT1 and AKT activation induced by CAV1 loss, implicating CAV1/NOS3 uncoupling and NOS3 dysregulation in the inflammatory phenotype. Exogenous IFN reduced CAV1 expression, activated STAT1 and AKT, and altered the cytoskeleton of PAECs, implicating these mechanisms in PAH associated with autoimmune and autoinflammatory diseases, as well as IFN therapy. CAV1 insufficiency elicits an IFN inflammatory response that results in a dysfunctional endothelial cell phenotype and targeting this pathway may reduce pathologic vascular remodeling in PAH.


Assuntos
Caveolina 1/genética , Endotélio Vascular/metabolismo , Hipertensão Pulmonar/metabolismo , Interferon Tipo I/metabolismo , Animais , Células Cultivadas , Endotélio Vascular/enzimologia , Endotélio Vascular/fisiopatologia , Inativação Gênica , Humanos , Hipertensão Pulmonar/fisiopatologia , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais
4.
Am J Physiol Lung Cell Mol Physiol ; 318(1): L98-L111, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31617731

RESUMO

Inflammatory cell infiltrates are a prominent feature of aberrant vascular remodeling in pulmonary arterial hypertension (PAH), suggesting that immune effector cells contribute to disease progression. Genome-wide blood expression profiling studies have attempted to better define this inflammatory component of PAH pathobiology but have been hampered by small sample sizes, methodological differences, and very little gene-level reproducibility. The current meta-analysis (seven studies; 156 PAH patients/110 healthy controls) was performed to assess the comparability of data across studies and to possibly derive a generalizable transcriptomic signature. Idiopathic (IPAH) compared with disease-associated PAH (APAH) displayed highly similar expression profiles with no differentially expressed genes, even after substantially relaxing selection stringency. In contrast, using a false discovery rate of ≤1% and I2 < 40% (low-to-moderate heterogeneity across studies) both IPAH and APAH differed markedly from healthy controls with the combined PAH cohort yielding 1,269 differentially expressed, unique gene transcripts. Bioinformatic analyses, including gene-set enrichment, which uses all available data independent of gene selection thresholds, identified interferon, mammalian target of rapamycin/p70S6K, stress kinase, and Toll-like receptor signaling as enriched mechanisms within the PAH gene signature. Enriched biological functions and diseases included tumorigenesis, autoimmunity, antiviral response, and cell death consistent with prevailing theories of PAH pathogenesis. Although otherwise indistinguishable, APAH (predominantly PAH due to systemic sclerosis) had a somewhat stronger interferon profile than IPAH. Meta-analysis defined a robust and generalizable transcriptomic signature in the blood of PAH patients that can help inform the identification of biomarkers and therapeutic targets.


Assuntos
Hipertensão Pulmonar Primária Familiar/genética , Hipertensão Arterial Pulmonar/genética , Transcriptoma/genética , Biomarcadores/metabolismo , Estudos de Casos e Controles , Feminino , Perfilação da Expressão Gênica/métodos , Humanos , Masculino , Reprodutibilidade dos Testes
5.
Monaldi Arch Chest Dis ; 89(1)2019 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-30968656

RESUMO

Associazione Riabilitatori dell'Insufficienza Respiratoria (ARIR) is pleased to announce a new editorial project by joining the Monaldi Archives of Chest Disease journal.


Assuntos
Publicações Periódicas como Assunto , Modalidades de Fisioterapia/tendências , Terapia Respiratória/tendências , Pesquisa Biomédica/organização & administração , Humanos , Itália , Modalidades de Fisioterapia/educação , Terapia Respiratória/educação
6.
J Infect Dis ; 218(10): 1631-1640, 2018 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-29868908

RESUMO

The major surface glycoprotein (Msg) is the most abundant surface protein among Pneumocystis species. Given that Msg is present on both the cyst and trophic forms of Pneumocystis and that dendritic cells play a critical role in initiating host immune responses, we undertook studies to examine activation of bone marrow-derived myeloid dendritic cells by Msg purified from Pneumocystis murina. Incubation of dendritic cells with Msg did not lead to increased expression of CD40, CD80, CD86, or major histocompatibility complex class II or to increased secretion of any of 10 cytokines. Microarray analysis identified very few differentially expressed genes. In contrast, lipopolysaccharide-activated dendritic cells had positive results of all of these assays. However, Msg did bind to mouse mannose macrophage receptor and human DC-SIGN, 2 C-type lectins expressed by dendritic cells that are important in recognition of pathogen-associated high-mannose glycoproteins. Deglycosylation of Msg demonstrated that this binding was dependent on glycosylation. These studies suggest that Pneumocystis has developed a mechanism to avoid activation of dendritic cells, potentially by the previously identified loss of genes that are responsible for the high level of protein mannosylation found in other fungi.


Assuntos
Células Dendríticas/efeitos dos fármacos , Proteínas Fúngicas/farmacologia , Glicoproteínas de Membrana/farmacologia , Pneumocystis/química , Animais , Células Cultivadas , Citocinas/análise , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL
7.
J Biol Chem ; 291(45): 23628-23644, 2016 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-27650495

RESUMO

Glucocorticoids are commonly used to treat inflammatory disorders. The glucocorticoid receptor (GR) can tether to inflammatory transcription factor complexes, such as NFκB and AP-1, and trans-repress the transcription of cytokines, chemokines, and adhesion molecules. In contrast, aldosterone and the mineralocorticoid receptor (MR) primarily promote cardiovascular inflammation by incompletely understood mechanisms. Although MR has been shown to weakly repress NFκB, its role in modulating AP-1 has not been established. Here, the effects of GR and MR on NFκB and AP-1 signaling were directly compared using a variety of ligands, two different AP-1 consensus sequences, GR and MR DNA-binding domain mutants, and siRNA knockdown or overexpression of core AP-1 family members. Both GR and MR repressed an NFκB reporter without influencing p65 or p50 binding to DNA. Likewise, neither GR nor MR affected AP-1 binding, but repression or activation of AP-1 reporters occurred in a ligand-, AP-1 consensus sequence-, and AP-1 family member-specific manner. Notably, aldosterone interactions with both GR and MR demonstrated a potential to activate AP-1. DNA-binding domain mutations that eliminated the ability of GR and MR to cis-activate a hormone response element-driven reporter variably affected the strength and polarity of these responses. Importantly, MR modulation of NFκB and AP-1 signaling was consistent with a trans-mechanism, and AP-1 effects were confirmed for specific gene targets in primary human cells. Steroid nuclear receptor trans-effects on inflammatory signaling are context-dependent and influenced by nuclear receptor conformation, DNA sequence, and the expression of heterologous binding partners. Aldosterone activation of AP-1 may contribute to its proinflammatory effects in the vasculature.


Assuntos
NF-kappa B/imunologia , Receptores de Glucocorticoides/imunologia , Receptores de Mineralocorticoides/imunologia , Transdução de Sinais , Fator de Transcrição AP-1/imunologia , Sequência de Aminoácidos , Sequência de Bases , DNA/química , Expressão Gênica , Regulação da Expressão Gênica , Células HEK293 , Humanos , Inflamação/genética , Inflamação/imunologia , Mutação , Domínios Proteicos , Receptores de Glucocorticoides/química , Receptores de Glucocorticoides/genética , Receptores de Mineralocorticoides/química , Receptores de Mineralocorticoides/genética
8.
Crit Care ; 20(1): 206, 2016 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-27368279

RESUMO

BACKGROUND: Dexmedetomidine can be used for sedation of mechanically ventilated patients and has minor respiratory effects. The aim of this study was to compare the incidence of patient-ventilator dyssynchronies during sedation with dexmedetomidine or propofol. METHODS: We conducted a multicentre, prospective, open-label, randomised clinical trial, comparing dexmedetomidine with standard propofol sedation at three intensive care units of university hospitals in Italy. Twenty difficult-to-wean patients for whom the first weaning trial had failed and who were on pressure support ventilation were randomised to receive sedation with either dexmedetomidine or propofol at a similar level of sedation (Richmond Agitation-Sedation Scale [RASS] score +1 to -2). The asynchrony index (AI) was calculated using tracings of airflow, airway pressure and electrical activity of the diaphragm sampled at 0, 0.5, 1, 2, 6, 12, 18 and 24 h. RESULTS: The mean AI was lower with dexmedetomidine than with propofol from 2 h onwards, although the two groups significantly differed only at 12 h (2.68 % vs 9.10 %, p < 0.05). No further difference was observed at 18 and 24 h. CONCLUSIONS: When sedation with propofol and dexmedetomidine was compared at similar RASS scores of patients in whom first weaning trial had failed, the AI was lower with dexmedetomidine than with propofol, and this difference was statistically significant at 12 h. These results suggest that sedation with dexmedetomidine may offer some advantages in terms of patient-ventilator synchrony.


Assuntos
Dexmedetomidina/efeitos adversos , Propofol/efeitos adversos , Desmame do Respirador/normas , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Dexmedetomidina/uso terapêutico , Feminino , Humanos , Hipnóticos e Sedativos/efeitos adversos , Hipnóticos e Sedativos/uso terapêutico , Unidades de Terapia Intensiva/organização & administração , Itália , Masculino , Pessoa de Meia-Idade , Propofol/uso terapêutico , Estudos Prospectivos , Respiração Artificial/métodos
9.
J Infect Dis ; 214(5): 782-91, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27324243

RESUMO

ß-glucans, which can activate innate immune responses, are a major component in the cell wall of the cyst form of Pneumocystis In the current study, we examined whether ß-1,3-glucans are masked by surface proteins in Pneumocystis and what role ß-glucans play in Pneumocystis-associated inflammation. For 3 species, including Pneumocystis jirovecii, which causes Pneumocystis pneumonia in humans, Pneumocystis carinii, and Pneumocystis murina, ß-1,3-glucans were masked in most organisms, as demonstrated by increased exposure following trypsin treatment. Using quantitative polymerase chain reaction and microarray techniques, we demonstrated in a mouse model of Pneumocystis pneumonia that treatment with caspofungin, an inhibitor of ß-1,3-glucan synthesis, for 21 days decreased expression of a broad panel of inflammatory markers, including interferon γ, tumor necrosis factor α, interleukin 1ß, interleukin 6, and multiple chemokines/chemokine ligands. Thus, ß-glucans in Pneumocystis cysts are largely masked, which likely decreases innate immune activation; this mechanism presumably was developed for interactions with immunocompetent hosts, in whom organism loads are substantially lower. In immunosuppressed hosts with a high organism burden, organism death and release of glucans appears to be an important contributor to deleterious host inflammatory responses.


Assuntos
Pneumocystis/imunologia , Pneumonia por Pneumocystis/patologia , Pneumonia/patologia , beta-Glucanas/imunologia , Animais , Antifúngicos/administração & dosagem , Caspofungina , Citocinas/análise , Modelos Animais de Doenças , Equinocandinas/administração & dosagem , Lipopeptídeos/administração & dosagem , Camundongos Knockout , Análise em Microsséries , Pneumonia por Pneumocystis/microbiologia , Reação em Cadeia da Polimerase em Tempo Real
10.
Am J Physiol Lung Cell Mol Physiol ; 310(2): L187-201, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26589479

RESUMO

A proliferative endothelial cell phenotype, inflammation, and pulmonary vascular remodeling are prominent features of pulmonary arterial hypertension (PAH). Bone morphogenetic protein type II receptor (BMPR2) loss-of-function is the most common cause of heritable PAH and has been closely linked to the formation of pathological plexiform lesions. Although some BMPR2 mutations leave ligand-dependent responses intact, the disruption of ligand-independent, noncanonical functions are universal among PAH-associated BMPR2 genotypes, but incompletely understood. This study examined the noncanonical signaling consequences of BMPR2 silencing in human pulmonary artery endothelial cells to identify potential therapeutic targets. BMPR2 siRNA silencing resulted in a proliferative, promigratory pulmonary artery endothelial cell phenotype and disruption of cytoskeletal architecture. Expression profiling closely reflected these phenotypic changes. Gene set enrichment and promoter analyses, as well as the differential expression of pathway components identified Ras/Raf/ERK signaling as an important consequence of BMPR2 silencing. Raf family members and ERK1/2 were constitutively activated after BMPR2 knockdown. Two Raf inhibitors, sorafenib and AZ628, and low-dose nintedanib, a triple receptor tyrosine kinase inhibitor upstream from Ras, reversed the abnormal proliferation and hypermotility of BMPR2 deficiency. Inhibition of dysregulated Ras/Raf/ERK signaling may be useful in reversing vascular remodeling in PAH.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Proliferação de Células , Células Endoteliais/citologia , Hipertensão Pulmonar/metabolismo , Pulmão/metabolismo , Artéria Pulmonar/metabolismo , Adulto , Idoso , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hipertensão Pulmonar Primária Familiar/metabolismo , Feminino , Inativação Gênica , Humanos , Pulmão/patologia , Masculino , Pessoa de Meia-Idade , Mutação/genética , Fenótipo , RNA Interferente Pequeno/genética , Transdução de Sinais/fisiologia , Remodelação Vascular/genética , Adulto Jovem , Quinases raf/metabolismo
11.
J Biol Chem ; 290(32): 19544-57, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26105050

RESUMO

Peroxisome proliferator-activated receptor γ (PPARγ) ligands have been widely used to treat type 2 diabetes mellitus. However, knowledge of PPARγ signaling remains incomplete. In addition to PPARγ, these drugs also activate G protein-coupled receptor 40 (GPR40), a Gαq-coupled free fatty acid receptor linked to MAPK networks and glucose homeostasis. Notably, p38 MAPK activation has been implicated in PPARγ signaling. Here, rosiglitazone (RGZ) activation of GPR40 and p38 MAPK was found to boost PPARγ-induced gene transcription in human endothelium. Inhibition or knockdown of p38 MAPK or expression of a dominant negative (DN) p38 MAPK mutant blunted RGZ-induced PPARγ DNA binding and reporter activity in EA.hy926 human endothelial cells. GPR40 inhibition or knockdown, or expression of a DN-Gαq mutant likewise blocked activation of both p38 MAPK and PPARγ reporters. Importantly, RGZ induction of PPARγ target genes in primary human pulmonary artery endothelial cells (PAECs) was suppressed by knockdown of either p38 MAPK or GPR40. GPR40/PPARγ signal transduction was dependent on p38 MAPK activation and induction of PPARγ co-activator-1 (PGC1α). Silencing of p38 MAPK or GPR40 abolished the ability of RGZ to induce phosphorylation and expression of PGC1α in PAECs. Knockdown of PGC1α, its essential activator SIRT1, or its binding partner/co-activator EP300 inhibited RGZ induction of PPARγ-regulated genes in PAECs. RGZ/GPR40/p38 MAPK signaling also led to EP300 phosphorylation, an event that enhances PPARγ target gene transcription. Thus, GPR40 and PPARγ can function as an integrated two-receptor signal transduction pathway, a finding with implications for rational drug development.


Assuntos
Células Endoteliais/metabolismo , PPAR gama/metabolismo , Receptor Cross-Talk , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Linhagem Celular , DNA/genética , DNA/metabolismo , Proteína p300 Associada a E1A/genética , Proteína p300 Associada a E1A/metabolismo , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Regulação da Expressão Gênica , Genes Reporter , Humanos , Hipoglicemiantes/farmacologia , Ligantes , Luciferases/genética , Luciferases/metabolismo , PPAR gama/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Pioglitazona , Cultura Primária de Células , Ligação Proteica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores Acoplados a Proteínas G/genética , Rosiglitazona , Sirtuína 1/genética , Sirtuína 1/metabolismo , Tiazolidinedionas/farmacologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/genética
12.
Thromb Haemost ; 112(4): 770-80, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25057108

RESUMO

Accurately detecting circulating endothelial cells (CECs) is important since their enumeration has been proposed as a biomarker to measure injury to the vascular endothelium. However, there is no single methodology for determining CECs in blood, making comparison across studies difficult. Many methods for detecting CECs rely on characteristic cell surface markers and cell viability indicators, but lack secondary validation. Here, a CEC population in healthy adult human subjects was identified by flow cytometry as CD45-, CD34dim that is comparable to a previously described CD45-, CD31bright population. In addition, nuclear staining with 7-aminoactinomycin D (7-AAD) was employed as a standard technique to exclude dead cells. Unexpectedly, the CD45-, CD34dim, 7-AAD- CECs lacked surface detectable CD146, a commonly used marker of CECs. Furthermore, light microscopy revealed this cell population to be composed primarily of large cells without a clearly defined nucleus. Nevertheless, immunostains still demonstrated the presence of the lectin Ulex europaeus and von Willebrand factor. Ultramicro analytical immunochemistry assays for the endothelial cell proteins CD31, CD34, CD62E, CD105, CD141, CD144 and vWF indicated these cells possess an endothelial phenotype. However, only a small amount of RNA, which was mostly degraded, could be isolated from these cells. Thus the majority of CECs in healthy individuals as defined by CD45-, CD34dim, and 7-AAD- have shed their CD146 surface marker and are senescent cells without an identifiable nucleus and lacking RNA of sufficient quantity and quality for transcriptomal analysis. This study highlights the importance of secondary validation of CEC identification.


Assuntos
Antígenos CD34/metabolismo , Separação Celular , Células Endoteliais/citologia , Antígenos Comuns de Leucócito/metabolismo , Adulto , Antígeno CD146/metabolismo , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Sobrevivência Celular , Dactinomicina/análogos & derivados , Dactinomicina/sangue , Citometria de Fluxo , Células Endoteliais da Veia Umbilical Humana , Humanos , Indóis/química , Leucócitos Mononucleares/citologia , Microscopia , Microscopia de Fluorescência , Pessoa de Meia-Idade , Fenótipo , Projetos Piloto , Lectinas de Plantas/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , RNA/metabolismo , Fator de von Willebrand/metabolismo
13.
PLoS One ; 9(2): e88756, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24551153

RESUMO

BACKGROUND: Bacterial superantigens are virulence factors that cause toxic shock syndrome. Here, the genome-wide, temporal response of mice to lethal intranasal staphylococcal enterotoxin B (SEB) challenge was investigated in six tissues. RESULTS: The earliest responses and largest number of affected genes occurred in peripheral blood mononuclear cells (PBMC), spleen, and lung tissues with the highest content of both T-cells and monocyte/macrophages, the direct cellular targets of SEB. In contrast, the response of liver, kidney, and heart was delayed and involved fewer genes, but revealed a dominant genetic program that was seen in all 6 tissues. Many of the 85 uniquely annotated transcripts participating in this shared genomic response have not been previously linked to SEB. Nine of the 85 genes were subsequently confirmed by RT-PCR in every tissue/organ at 24 h. These 85 transcripts, up-regulated in all tissues, annotated to the interferon (IFN)/antiviral-response and included genes belonging to the DNA/RNA sensing system, DNA damage repair, the immunoproteasome, and the ER/metabolic stress-response and apoptosis pathways. Overall, this shared program was identified as a type I and II interferon (IFN)-response and the promoters of these genes were highly enriched for IFN regulatory matrices. Several genes whose secreted products induce the IFN pathway were up-regulated at early time points in PBMCs, spleen, and/or lung. Furthermore, IFN regulatory factors including Irf1, Irf7 and Irf8, and Zbp1, a DNA sensor/transcription factor that can directly elicit an IFN innate immune response, participated in this host-wide SEB signature. CONCLUSION: Global gene-expression changes across multiple organs implicated a host-wide IFN-response in SEB-induced death. Therapies aimed at IFN-associated innate immunity may improve outcome in toxic shock syndromes.


Assuntos
Imunidade Inata/genética , Fatores Reguladores de Interferon/genética , Choque Séptico/genética , Transcriptoma/imunologia , Administração Intranasal , Animais , Enterotoxinas , Regulação da Expressão Gênica , Estudo de Associação Genômica Ampla , Injeções Intraperitoneais , Fatores Reguladores de Interferon/imunologia , Rim/imunologia , Rim/patologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/patologia , Fígado/imunologia , Fígado/patologia , Pulmão/imunologia , Pulmão/patologia , Masculino , Camundongos , Miocárdio/imunologia , Miocárdio/patologia , Regiões Promotoras Genéticas , Choque Séptico/induzido quimicamente , Choque Séptico/imunologia , Choque Séptico/mortalidade , Baço/imunologia , Baço/patologia , Análise de Sobrevida
14.
J Exp Med ; 210(10): 1961-76, 2013 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-23999499

RESUMO

Dendritic cells (DCs) comprise distinct populations with specialized immune-regulatory functions. However, the environmental factors that determine the differentiation of these subsets remain poorly defined. Here, we report that retinoic acid (RA), a vitamin A derivative, controls the homeostasis of pre-DC (precursor of DC)-derived splenic CD11b(+)CD8α(-)Esam(high) DCs and the developmentally related CD11b(+)CD103(+) subset within the gut. Whereas mice deprived of RA signaling significantly lost both of these populations, neither pre-DC-derived CD11b(-)CD8α(+) and CD11b(-)CD103(+) nor monocyte-derived CD11b(+)CD8α(-)Esam(low) or CD11b(+)CD103(-) DC populations were deficient. In fate-tracking experiments, transfer of pre-DCs into RA-supplemented hosts resulted in near complete conversion of these cells into the CD11b(+)CD8α(-) subset, whereas transfer into vitamin A-deficient (VAD) hosts caused diversion to the CD11b(-)CD8α(+) lineage. As vitamin A is an essential nutrient, we evaluated retinoid levels in mice and humans after radiation-induced mucosal injury and found this conditioning led to an acute VAD state. Consequently, radiation led to a selective loss of both RA-dependent DC subsets and impaired class II-restricted auto and antitumor immunity that could be rescued by supplemental RA. These findings establish a critical role for RA in regulating the homeostasis of pre-DC-derived DC subsets and have implications for the management of patients with immune deficiencies resulting from malnutrition and irradiation.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Homeostase/imunologia , Tretinoína/metabolismo , Animais , Diferenciação Celular/imunologia , Proliferação de Células , Sobrevivência Celular , Células Dendríticas/citologia , Células Dendríticas/efeitos da radiação , Feminino , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Imunofenotipagem , Mucosa Intestinal/metabolismo , Intestinos/imunologia , Intestinos/efeitos da radiação , Camundongos , Neoplasias/imunologia , Neoplasias/metabolismo , Especificidade de Órgãos/imunologia , Fenótipo , Receptores do Ácido Retinoico/metabolismo , Transdução de Sinais , Baço/imunologia , Baço/metabolismo , Baço/efeitos da radiação , Vitamina A/metabolismo , Irradiação Corporal Total/efeitos adversos
15.
Free Radic Biol Med ; 53(12): 2229-39, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23099417

RESUMO

Plasma hemoglobin (Hb) released during intravascular hemolysis has been associated with numerous deleterious effects that may stem from increased nitric oxide (NO) scavenging, but has also been associated with reactive oxygen species generation and platelet activation. Therapies that convert plasma oxyHb to metHb, or metHb to iron-nitrosyl Hb, could be beneficial because these species do not scavenge NO. In this study, we investigated the effects of Angeli's salt (AS; sodium α-oxyhyponitrite, Na2N2O3), a nitroxyl (HNO) and nitrite (NO2(-)) donor, on plasma Hb oxidation and formation of iron-nitrosyl Hb from metHb and on the vasoactivity of plasma Hb. We hypothesized that AS could ameliorate hemolysis-associated pathology via its preferential reactivity with plasma Hb, as opposed to red-cell-encapsulated Hb, and through its intrinsic vasodilatory activity. To test this hypothesis, we infused (n=3 per group) (1) cell-free Hb and AS, (2) cell-free Hb+0.9% NaCl, (3) AS+3% albumin, and (4) 3% albumin+0.9% NaCl (colloid controls for Hb and AS, respectively) in a canine model. Co-infusion of AS and cell-free Hb led to preferential conversion of plasma Hb to metHb, but the extent of conversion was lower than anticipated based on the in vivo concentration of AS relative to plasma Hb. This lower metHb yield was probably due to reactions of nitroxyl-derived AS with plasma components such as thiol-containing compounds. From a physiological and therapeutic standpoint, the infusion of Hb alone led to significant increases in mean arterial pressure (p=0.03) and systemic vascular resistance index (p=0.01) compared to controls. Infusion of AS alone led to significant decreases in these parameters and co-infusion of AS along with Hb had an additive effect in reversing the effects of Hb alone on the systemic circulation. Interestingly, in the pulmonary system, the decrease in pressure when AS was added to Hb was significantly less than would have been expected compared to the effects of Hb and AS alone, suggesting that inactivation of scavenging with AS reduced the direct vasodilatory effects of AS on the vasculature. We also found that AS reduced platelet activation when administered to whole blood in vitro. These data suggest that AS-like compounds could serve as therapeutic agents to counteract the negative vasoconstrictive consequences of hemolysis that occur in hemolytic anemias, transfusion of stored blood, and other diseases. Increases in metHb in the red blood cell, the potential of AS for neurotoxicity, and hypotension would need to be carefully monitored in a clinical trial.


Assuntos
Metemoglobina/metabolismo , Nitritos/farmacologia , Vasodilatadores/farmacologia , Animais , Plaquetas/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Cães , Eritrócitos/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Leucócitos/efeitos dos fármacos , Oxirredução , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/fisiopatologia , Vasoconstrição
16.
Immunity ; 35(6): 972-85, 2011 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-22177921

RESUMO

Th17 cells have been described as short lived, but this view is at odds with their capacity to trigger protracted damage to normal and transformed tissues. We report that Th17 cells, despite displaying low expression of CD27 and other phenotypic markers of terminal differentiation, efficiently eradicated tumors and caused autoimmunity, were long lived, and maintained a core molecular signature resembling early memory CD8(+) cells with stem cell-like properties. In addition, we found that Th17 cells had high expression of Tcf7, a direct target of the Wnt and ß-catenin signaling axis, and accumulated ß-catenin, a feature observed in stem cells. In vivo, Th17 cells gave rise to Th1-like effector cell progeny and also self-renewed and persisted as IL-17A-secreting cells. Multipotency was required for Th17 cell-mediated tumor eradication because effector cells deficient in IFN-γ or IL-17A had impaired activity. Thus, Th17 cells are not always short lived and are a less-differentiated subset capable of superior persistence and functionality.


Assuntos
Células-Tronco/metabolismo , Células Th17/imunologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Sobrevivência Celular/genética , Perfilação da Expressão Gênica , Interleucina-17/biossíntese , Camundongos , Camundongos Transgênicos , Neoplasias/imunologia , Células-Tronco/citologia , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Células Th1/citologia , Células Th1/imunologia , Células Th17/citologia , Células Th17/metabolismo , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo
17.
J Trauma ; 68(6): 1317-27, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20068480

RESUMO

BACKGROUND: Supporting its therapeutic application in sepsis, p38 mitogen-activated protein kinase (MAPK) inhibition decreases cardiopulmonary injury and lethality with lipopolysaccharide challenge. However, only one preclinical study has reported the survival effects of a p38 inhibitor (SB203580, 100 mg/kg) during infection. We therefore tested SB203580 in mice (n = 763) challenged with intratracheal Escherichia coli and treated with antibiotics and fluids. METHODS AND RESULTS: Compared with placebo, high dose SB203580 (100 mg/kg) pretreatment increased the hazards ratio of death (95% confidence interval) (3.6 [2.1, 6.1], p < 0.0001). Decreasing doses (10, 1, or 0.1 mg/kg) went from being harmful to having no significant effect (p < 0.0001 for the effect of decreasing dose). At 48 hours, but not 24 hours after E. coli, high and low dose SB203580 pretreatment decreased cardiac phosphorylated p38 MAPK levels and improved cardiac output either (p

Assuntos
Inibidores Enzimáticos/farmacologia , Infecções por Escherichia coli/tratamento farmacológico , Imidazóis/farmacologia , Pneumonia/tratamento farmacológico , Piridinas/farmacologia , Análise de Variância , Animais , Antibacterianos/farmacologia , Débito Cardíaco/efeitos dos fármacos , Ecocardiografia , Inibidores Enzimáticos/administração & dosagem , Ensaio de Imunoadsorção Enzimática , Infecções por Escherichia coli/fisiopatologia , Hidratação , Imidazóis/administração & dosagem , Lipopolissacarídeos/farmacologia , Lesão Pulmonar/fisiopatologia , Camundongos , Placebos , Pneumonia/fisiopatologia , Modelos de Riscos Proporcionais , Piridinas/administração & dosagem , Taxa de Sobrevida
18.
PLoS One ; 4(12): e8139, 2009 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-19956541

RESUMO

Carbon monoxide (CO) is an endogenous messenger that suppresses inflammation, modulates apoptosis and promotes vascular remodeling. Here, microarrays were employed to globally characterize the CO (250 ppm) suppression of early (1 h) LPS-induced inflammation in human monocytic THP-1 cells. CO suppressed 79 of 101 immediate-early genes induced by LPS; 19% (15/79) were transcription factors and most others were cytokines, chemokines and immune response genes. The prototypic effects of CO on transcription and protein production occurred early but decreased rapidly. CO activated p38 MAPK, ERK1/2 and Akt and caused an early and transitory delay in LPS-induced JNK activation. However, selective inhibitors of these kinases failed to block CO suppression of LPS-induced IL-1beta, an inflammation marker. Of CO-suppressed genes, 81% (64/79) were found to have promoters with putative NF-kappaB binding sites. CO was subsequently shown to block LPS-induced phosphorylation and degradation of IkappaBalpha in human monocytes, thereby inhibiting NF-kappaB signal transduction. CO broadly suppresses the initial inflammatory response of human monocytes to LPS by reshaping proximal events in TLR4 signal transduction such as stress kinase responses and early NF-kappaB activation. These rapid, but transient effects of CO may have therapeutic applications in acute pulmonary and vascular injury.


Assuntos
Monóxido de Carbono/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Monócitos/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Humanos , Proteínas I-kappa B/metabolismo , Interleucina-1beta/biossíntese , Interleucina-1beta/genética , Interleucina-8/genética , Interleucina-8/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Monócitos/enzimologia , Inibidor de NF-kappaB alfa , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reprodutibilidade dos Testes , Fatores de Tempo , Receptor 4 Toll-Like/genética
19.
Curr Opin Crit Care ; 15(4): 342-8, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19542885

RESUMO

PURPOSE OF REVIEW: To discuss the recent literature concerning the significance of respiratory complications as a determining factor in postoperative complications after major surgery. Although many studies have identified risk factors focusing on the prevention of respiratory complications, these complications continue to be a significant cause of morbidity and mortality. Here, we will examine the diagnosis, contributing factors, consequences, and early treatment of respiratory complications. RECENT FINDINGS: General anesthesia and surgery are the main causes of postoperative respiratory complications. Atelectasis, a common respiratory complication, may contribute to pneumonia and acute respiratory failure. Recently, it has been shown that activation of abdominal muscles during the induction of anesthesia contributes to a reduction of lung capacity, leading to a higher degree of atelectasis. Additionally, long-term mortality at 5 and 10 years has recently been shown to remain significantly increased in patients with respiratory complications. Prevention or early therapy of respiratory complications may, therefore, be beneficial in improving outcome in postoperative patients. SUMMARY: Postoperative respiratory complications may have significant deleterious consequences. Increasing the understanding of the underlying causes of respiratory complications and developing early treatment strategies will likely provide improved benefits. To date, early treatment with prophylactic or therapeutic continuous positive airway pressure has proved beneficial in an abdominal surgical patient population; however, the efficacy in a general population remains unclear.


Assuntos
Complicações Pós-Operatórias , Insuficiência Respiratória/etiologia , Pressão Positiva Contínua nas Vias Aéreas , Humanos , Hipóxia/etiologia , Atelectasia Pulmonar/etiologia , Insuficiência Respiratória/fisiopatologia
20.
Anesth Analg ; 109(1): 164-7, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19439687

RESUMO

Noninvasive ventilation (NIV) has been widely used to decrease the complications associated with tracheal intubation in mechanically ventilated patients with neuromuscular diseases in acute respiratory failure. However, nasal ulcerations might occur when masks are used as an interface. Helmet ventilation is a possible option in this case. We describe two patients with acute respiratory failure due to Duchenne muscular dystrophy who developed nasal bridge skin necrosis during NIV. Helmet pressure support ventilation caused significant patient-ventilator asynchrony, leading to NIV intolerance. Thus, biphasic positive airway pressure delivered by helmet was applied, which improved gas exchange and patient-ventilator interaction, allowing successful NIV.


Assuntos
Dispositivos de Proteção da Cabeça , Doenças Neuromusculares/terapia , Respiração Artificial/instrumentação , Síndrome do Desconforto Respiratório/terapia , Úlcera Cutânea/prevenção & controle , Adolescente , Humanos , Masculino , Máscaras/efeitos adversos , Doenças Neuromusculares/complicações , Doenças Neuromusculares/fisiopatologia , Respiração Artificial/métodos , Síndrome do Desconforto Respiratório/etiologia , Úlcera Cutânea/etiologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...