Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Cell Biol Int ; 46(4): 660-670, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35066972

RESUMO

The predominance of cardiovascular diseases among men compared to premenopausal women has been attributed to testosterone, which is implicated in vascular remodeling. Molecular mechanisms underlying its role have not been clarified but oxidative stress-induced inflammation may be important. We therefore investigated in vitro the effects of testosterone and dihydrotestosterone, (a nonaromatized androgen), on redox homeostasis in absence (basal conditions) and after corticotropin-releasing hormone-induced pro-oxidant action in macroendothelial cells. More specifically, we explored their role on well-established antioxidant enzymes activity, namely endothelial nitric oxide synthase, superoxide dismutase, catalase, and glutathione. We observed that both androgens significantly increased the intracellular reactive oxygen species levels, endothelial nitric oxide synthase activity, nitric oxide concentration as well as superoxide dismutase activity and decreased catalase activity. These effects of Testosterone and DHT were reversed in the presence of the androgen receptor antagonist, flutamide. Moreover, testosterone and dihydrotestosterone similarly enhanced the stimulatory effect of corticotropin-releasing hormone on intracellular reactive oxygen species levels and superoxide dismutase activity but did not influence the inhibitory effect on endothelial nitric oxide synthase activity, nitric oxide release and catalase activity. Finally, androgens did not have a detectable effect on glutathione levels or the glutathione/glutathione plus glutathione disulfide ratio. Our results reveal that testosterone and DHT rise the intracellular redox threshold of the endothelial cell and increases NO synthesis. These findings suggest that the action of testosterone is affected by the redox status of the endothelium and help to explain its controversial effects on the cardiovascular system.


Assuntos
Di-Hidrotestosterona , Testosterona , Di-Hidrotestosterona/farmacologia , Endotélio , Endotélio Vascular , Feminino , Homeostase , Humanos , Masculino , Óxido Nítrico , Oxirredução , Testosterona/farmacologia
2.
J Cell Sci ; 132(7)2019 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-30962349

RESUMO

Hypoxia-inducible factor 2 (HIF-2) is a principal component of the cellular response to oxygen deprivation (hypoxia). Its inducible subunit, HIF-2α (also known as EPAS1), is controlled by oxygen-dependent as well as oxygen-independent mechanisms, such as phosphorylation. We show here that HIF-2α is phosphorylated under hypoxia (1% O2) by extracellular signal-regulated protein kinases 1 and 2 (ERK1/2; also known as MAPK3 and MAPK1, respectively) at serine residue 672, as identified by in vitro phosphorylation assays. Mutation of this site to an alanine residue or inhibition of the ERK1/2 pathway decreases HIF-2 transcriptional activity and causes HIF-2α to mislocalize to the cytoplasm without changing its protein expression levels. Localization, reporter gene and immunoprecipitation experiments further show that HIF-2α associates with the exportin chromosomal maintenance 1 (CRM1, also known as XPO1) in a phosphorylation-sensitive manner and identify two critical leucine residues as part of an atypical CRM1-dependent nuclear export signal (NES) neighboring serine 672. Inhibition of CRM1 or mutation of these residues restores nuclear accumulation and activity of HIF-2α lacking the ERK1/2-mediated modification. In summary, we reveal a novel regulatory mechanism of HIF-2, involving ERK1/2-dependent phosphorylation of HIF-2α, which controls its nucleocytoplasmic shuttling and the HIF-2 transcriptional activity.This article has an associated First Person interview with the first author of the paper.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Núcleo Celular/metabolismo , Carioferinas/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Transporte Ativo do Núcleo Celular , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Núcleo Celular/genética , Células HeLa , Humanos , Carioferinas/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Mutação , Fosforilação , Receptores Citoplasmáticos e Nucleares/genética , Serina/metabolismo , Proteína Exportina 1
3.
Biochem Biophys Res Commun ; 557: 143-150, 2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-33865222

RESUMO

Hypoxia-inducible factor 2 (HIF-2), is essential for cellular response to hypoxia and holds an important role in erythropoiesis, angiogenesis, tissue invasion and metastasis, thus, constituting an important therapeutic target. Maximal HIF-2 transcriptional activation requires HIF-2α phosphorylation by ERK1/2 that impairs its CRM1-mediated nuclear export. Herein, we reveal a novel interaction of HIF-2α with Reptin52, a multifunctional protein involved in cellular functions orchestrated both in the nucleus and the cytoplasm. HIF-2α and Reptin52 interact both in nuclear and cytoplasmic fractions, however, ERK1/2 pathway inactivation seems to favour their association in the cytoplasm. Notably, we demonstrate that Reptin52 reduces HIF-2 transcriptional activity, which results in decreased EPO secretion under hypoxia, by impairing HIF-2α stability via a non-canonical PHD-VHL-proteasome independent mechanism. This interaction represents a novel HIF-2 fine tuning mechanism that allows for distinct HIF1/2 isoforms regulation.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Transporte/metabolismo , DNA Helicases/metabolismo , Eritropoetina/metabolismo , Regulação da Expressão Gênica/genética , Sistema de Sinalização das MAP Quinases/genética , ATPases Associadas a Diversas Atividades Celulares/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteínas de Transporte/genética , Hipóxia Celular , Linhagem Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cromatografia Líquida , Citoplasma/genética , Citoplasma/metabolismo , DNA Helicases/genética , Eritropoese/genética , Eritropoetina/genética , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Complexo de Endopeptidases do Proteassoma/metabolismo , Estabilidade Proteica , RNA Interferente Pequeno , Reação em Cadeia da Polimerase em Tempo Real , Espectrometria de Massas em Tandem
4.
J Surg Res ; 234: 325-333, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30527492

RESUMO

BACKGROUND: The use of surgical meshes in ventral hernia repair has significantly reduced hernia recurrence rates. However, when placed intraperitoneally prosthetic materials can trigger the development of peritoneal adhesions. The present experimental study evaluated the combined icodextrin 4% and dimetindene maleate treatment in preventing peritoneal adhesion formation to polypropylene and titanium-coated polypropylene meshes. MATERIALS AND METHODS: Sixty female white rabbits were divided into four groups. A 2 × 2 cm piece of mesh was fixed to intact peritoneum in all animals through a midline laparotomy. A lightweight polypropylene mesh was implanted in groups 1 and 2 and a titanium-coated polypropylene mesh in groups 3 and 4. Groups 2 and 4 were treated, intraoperatively, with intravenous dimetindene maleate (0.1 mg/kg) and intraperitoneal solution of icodextrin 4% (20 mL/kg) and for the next 6 d with dimetindene maleate intramuscularly. The observation period lasted 15 d. Adhesion scores, percentage of mesh affected surface, tissue hydroxyproline levels, and tissue histopathology were examined. RESULTS: All animals in group 1 and 57% of animals in group 3 presented postoperative adhesions. The combination of antiadhesives significantly reduced the extent and severity of adhesions as well as the hydroxyproline levels in groups 2 and 4 compared with groups 1 and 3. On microscopic evaluation, animals in group 1 exhibited higher inflammation scores compared with group 2, whereas animals in groups 2 and 4 had better mesotheliazation compared with groups 1 and 3. CONCLUSIONS: The combined administration of icodextrin 4% and dimetindene maleate reduces the extent and severity of adhesions and may be successfully used to prevent adhesion formation after mesh intraperitoneal placement.


Assuntos
Dimetideno/administração & dosagem , Icodextrina/administração & dosagem , Polipropilenos/efeitos adversos , Complicações Pós-Operatórias/prevenção & controle , Substâncias Protetoras/administração & dosagem , Telas Cirúrgicas/efeitos adversos , Aderências Teciduais/prevenção & controle , Animais , Dimetideno/uso terapêutico , Quimioterapia Combinada , Feminino , Icodextrina/uso terapêutico , Injeções Intramusculares , Injeções Intraperitoneais , Injeções Intravenosas , Complicações Pós-Operatórias/etiologia , Substâncias Protetoras/uso terapêutico , Coelhos , Distribuição Aleatória , Resultado do Tratamento
5.
Cell Biol Int ; 43(12): 1407-1415, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31141240

RESUMO

Corticotropin-releasing hormone, which is the predominant regulator of neuroendocrine responses to stress, attenuates inflammation through stimulation of glucocorticoid release. Enhanced corticotropin-releasing hormone expression has been detected in inflammatory cells of the vascular endothelium, where it acts as a local regulator of endothelial redox homeostasis. Estrogens have beneficial effects on endothelial integrity and function, though the mechanism underlying their antioxidative effect remains as yet largely unknown. We therefore investigated the effect of 17ß-estradiol on pro-oxidant action of corticotropin-releasing hormone in vitro in macroendothelial cells, and, more specifically, the role of 17ß-estradiol on corticotropin-releasing hormone-induced activities/release of the antioxidant enzymes namely, endothelial nitric oxide synthase, superoxide dismutase, catalase, and glutathione. We observed that 17ß-estradiol abolished the stimulatory effect of corticotropin-releasing hormone on intracellular reactive oxygen species levels and counteracted its inhibitory effect on endothelial nitric oxide synthase activity and nitric oxide release. In addition, 17ß-estradiol significantly induced superoxide dismutase and catalase activity, an effect that was not significantly influenced by corticotropin-releasing hormone. Finally, 17ß-estradiol significantly increased glutathione levels and the glutathione/glutathione + glutathione disulfide ratio, an action that was partially blocked by corticotropin-releasing hormone. Our results reveal that 17ß-estradiol counterbalances corticotropin-releasing hormone-mediated pro-inflammatory action and thereby maintains the physiological threshold of the endothelial cell redox environment. These observations may be of importance, considering the protective role of estrogen in the development of atherosclerosis.

6.
J Cell Physiol ; 233(12): 9087-9098, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29968905

RESUMO

Angiogenesis is a key enabling feature of mammalian embryonic development and tumor progression, which provides oxygen and nutrients that are required for vessel growth and tumor cell growth, respectively. Hypoxia is a driver of this phenomenon and is considered to be one of the most potent initiators of angiogenesis both in vitro and in vivo through stabilization of the transcription factors, hypoxia-inducible factor-1 and -2 (HIF-1 and HIF-2). Although these proteins are highly homologous, emerging evidence suggests that they have unique transcriptional targets and differential impact on angiogenesis. Although HIF-1α is the best known and widely described isoform, recent studies suggest that HIF-2α is a critical regulator of physiological and pathophysiological angiogenesis and, at least, the similiarly important as HIF-1α. Indeed, HIF-2α has been shown to regulate multiple aspects of angiogenesis, including cell proliferation, migration, maturation of blood vessels, and metastasis. In this review, we focus on recent insights into HIF-2α expression, activation, and function under hypoxic and nonhypoxic conditions. We also summarize the current knowledge on the crosstalk between HIF-2 and angiogenesis, describing reported phenotypical changes of HIF-2α genetic models and HIF-2 target genes implicated in angiogenesis. Finally, we provide a survey of recent pharmacologic strategies to specifically target HIF-2 activity.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Hipóxia Celular/genética , Neovascularização Patológica/genética , Neovascularização Fisiológica/genética , Proliferação de Células/genética , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário/genética , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias/genética , Neoplasias/patologia , Neovascularização Patológica/patologia , Neovascularização Fisiológica/fisiologia , Oxigênio/metabolismo
7.
J Cell Sci ; 129(22): 4213-4226, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27686097

RESUMO

Hypoxia inducible factor 2 (HIF-2) is a transcriptional activator implicated in the cellular response to hypoxia. Regulation of its inducible subunit, HIF-2α (also known as EPAS1), involves post-translational modifications. Here, we demonstrate that casein kinase 1δ (CK1δ; also known as CSNK1D) phosphorylates HIF-2α at Ser383 and Thr528 in vitro We found that disruption of these phosphorylation sites, and silencing or chemical inhibition of CK1δ, reduced the expression of HIF-2 target genes and the secretion of erythropoietin (EPO) in two hepatic cancer cell lines, Huh7 and HepG2, without affecting the levels of HIF-2α protein expression. Furthermore, when CK1δ-dependent phosphorylation of HIF-2α was inhibited, we observed substantial cytoplasmic mislocalization of HIF-2α, which was reversed upon the addition of the nuclear protein export inhibitor leptomycin B. Taken together, these data suggest that CK1δ enhances EPO secretion from liver cancer cells under hypoxia by modifying HIF-2α and promoting its nuclear accumulation. This modification represents a new mechanism of HIF-2 regulation that might allow HIF isoforms to undertake differing functions.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Caseína Quinase Idelta/metabolismo , Eritropoetina/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/química , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Hipóxia Celular , Núcleo Celular/metabolismo , Inativação Gênica , Células HeLa , Células Hep G2 , Humanos , Carioferinas/metabolismo , Neoplasias Hepáticas/genética , Mutação/genética , Fosforilação , Ligação Proteica , Estabilidade Proteica , Receptores Citoplasmáticos e Nucleares/metabolismo , Transcrição Gênica , Fatores Estimuladores Upstream/metabolismo , Proteína Exportina 1
8.
Cell Biol Int ; 41(7): 769-778, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28418174

RESUMO

Tissue hypoxia affects gene expression through the hypoxia-inducible transcription factors, HIF-1 and HIF-2, in both physiological and pathological angiogenesis. Angiogenesis is a complex response of endothelial cells integrating cell proliferation, migration, tube formation, and their interaction with the extracellular matrix through integrin receptors. In this report, we studied the effect of hypoxia on the angiogenic functions of human microvascular endothelial cells (HMEC-1) as well as on expression of the angiogenic integrins αν ß3 , αν ß5 , and α5 ß1 . Exposure of HMEC-1 to hypoxia (1% O2 ) or to DMOG, a prolyl-4-hydroxylase inhibitor, caused significant reduction to their proliferation rate, whereas their migration ability toward laminin-1 or collagen IV and capillary-like tube formation were significantly enhanced. In addition, αv , ß1 , ß3 , and ß5 integrins expression was increased under hypoxia in HMEC-1, while α5 integrin was not affected. Both HIF-1 and HIF-2 protein expression and transcriptional activity were induced under hypoxia in HMEC-1. The knockdown of either HIF-1α or HIF-2α inhibited integrin ß3 hypoxic stimulation, suggesting a HIF-dependent induction of ß3 integrin in HMEC-1. Taken together, our results indicate that hypoxia transcriptionally up-regulates angiogenic integrins in microvascular endothelial cells along with promoting migration and tube formation of HMEC-1.


Assuntos
Hipóxia Celular/fisiologia , Movimento Celular/fisiologia , Endotélio Vascular/fisiologia , Integrinas/biossíntese , Integrinas/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Movimento Celular/genética , Proliferação de Células/fisiologia , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Humanos , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Integrinas/metabolismo , Neovascularização Fisiológica/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Regulação para Cima
9.
Biochim Biophys Acta ; 1833(6): 1378-87, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23458834

RESUMO

Hypoxia inducible factor-1 (HIF-1), a dimeric transcription factor of the bHLH-PAS family, is comprised of HIF-1α, which is inducible by hypoxia and ARNT or HIF-1ß, which is constitutively expressed. HIF-1 is involved in cellular homeostasis under hypoxia, in development and in several diseases affected by oxygen availability, particularly cancer. Since its expression is positively correlated with poor outcome prognosis for cancer patients, HIF-1 is a target for pharmaceutical therapy. We have previously shown that male germ cell Rac GTPase activating protein (MgcRacGAP), a regulator of Rho proteins which are principally involved in cytoskeletal organization, binds to HIF-1α and inhibits its transcriptional activity. In this work, we have explored the mechanism of the MgcRacGAP-mediated HIF-1 inactivation. We show that the Myo domain of MgcRacGAP, which is both necessary and sufficient for HIF-1 repression, binds to the PAS-B domain of HIF-1α. Furthermore MgcRacGAP competes with ARNT for binding to the HIF-1α PAS-B domain, as shown by in vitro binding pull down assays. In mammalian cells, ARNT overexpression can overcome the MgcRacGAP-mediated inhibition and MgcRacGAP binding to HIF-1α in vivo inhibits its dimerization with ARNT. We additionally present results indicating that MgcRacGAP binding to HIF-1α is specific, since it does not affect the transcriptional activity of HIF-2, a close evolutionary relative of HIF-1 also involved in hypoxia regulation and cancer. Our results reveal a new mechanism for HIF-1 transcriptional activity regulation, suggest a novel hypoxia-cytoskeleton link and provide new tools for selective HIF-1 inhibition.


Assuntos
Translocador Nuclear Receptor Aril Hidrocarboneto/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Regulação da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Hipóxia , Transcrição Gênica , Translocador Nuclear Receptor Aril Hidrocarboneto/genética , Western Blotting , Células Cultivadas , Citoesqueleto , Proteínas Ativadoras de GTPase/genética , Humanos , Imunoprecipitação , Ligação Proteica , Multimerização Proteica , Estrutura Terciária de Proteína , Ativação Transcricional
10.
J Cell Sci ; 125(Pt 14): 3485-93, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22467849

RESUMO

Adaptation to hypoxia involves hypoxia-inducible transcription factors (HIFs) and requires reprogramming of cellular metabolism that is essential during both physiological and pathological processes. In contrast to the established role of HIF-1 in glucose metabolism, the involvement of HIFs and the molecular mechanisms concerning the effects of hypoxia on lipid metabolism are poorly characterized. Here, we report that exposure of human cells to hypoxia causes accumulation of triglycerides and lipid droplets. This is accompanied by induction of lipin 1, a phosphatidate phosphatase isoform that catalyzes the penultimate step in triglyceride biosynthesis, whereas lipin 2 remains unaffected. Hypoxic upregulation of lipin 1 expression involves predominantly HIF-1, which binds to a single distal hypoxia-responsive element in the lipin 1 gene promoter and causes its activation under low oxygen conditions. Accumulation of hypoxic triglycerides or lipid droplets can be blocked by siRNA-mediated silencing of lipin 1 expression or kaempferol-mediated inhibition of HIF-1. We conclude that direct control of lipin 1 transcription by HIF-1 is an important regulatory feature of lipid metabolism and its adaptation to hypoxia.


Assuntos
Hipóxia Celular/fisiologia , Fator 1 Induzível por Hipóxia/metabolismo , Fosfatidato Fosfatase/biossíntese , Triglicerídeos/metabolismo , Hipóxia Celular/genética , Linhagem Celular Tumoral , Células HeLa , Humanos , Fator 1 Induzível por Hipóxia/genética , Fosfatidato Fosfatase/genética , Regiões Promotoras Genéticas , Triglicerídeos/biossíntese , Triglicerídeos/genética , Regulação para Cima
11.
Invest New Drugs ; 31(5): 1169-81, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23645448

RESUMO

Development of drug resistance after standard chemotherapy for glioblastoma multiforme (GBM) with temozolomide (TMZ) is associated with poor prognosis of GBM patients and is at least partially mediated by a direct DNA repair pathway involving O6-methylguanine methyltransferase (MGMT). This enzyme is under post-translational control by a multisubunit proteolytic cellular machinery, the 26S proteasome. Inhibition of the proteasome by bortezomib (BZ), a boronic acid dipeptide already in clinical use for the treatment of myeloma, has been demonstrated to induce growth arrest and apoptosis in GBM cells. In this study we investigated the effect of sequential treatment with BZ and TMZ on cell proliferation-viability and apoptosis of the human T98G and U87 GBM cell lines. We also tested for an effect of treatment on MGMT expression and important upstream regulators of the latter, including nuclear factor kappa B (NFκB), p44/42 mitogen-activated protein kinase (MAPK), p53, signal transducer and activator of transcription 3 (STAT3) and hypoxia-inducible factor 1α (HIF-1α). The sequence of drug administration for maximal cytotoxicity favored BZ prior to TMZ in T98G cells while the opposite was the case for U87 cells. Maximal efficacy was associated with downregulation of MGMT, reduced IκBα-mediated proteasome-dependent nuclear accumulation of NFκB, attenuation of p44/42 MAPK, AKT and STAT3 activation, and stabilization of p53 and inactive HIF-1α. Collectively, these results suggest that proteasome inhibition by BZ overcomes MGMT-mediated GBM chemoresistance, with scheduling of administration being critical for obtaining the maximal tumoricidal effect of combination with TMZ.


Assuntos
Antineoplásicos/administração & dosagem , Ácidos Borônicos/administração & dosagem , Dacarbazina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , O(6)-Metilguanina-DNA Metiltransferase/antagonistas & inibidores , Pirazinas/administração & dosagem , Bortezomib , Linhagem Celular Tumoral , Dacarbazina/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/fisiologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Transcrição STAT3/metabolismo , Temozolomida , Proteína Supressora de Tumor p53/metabolismo
12.
Sleep Breath ; 16(3): 703-8, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21811879

RESUMO

INTRODUCTION: Obstructive sleep apnea (OSA) in children has been associated with systemic inflammation and oxidative stress. Limited evidence indicates that pediatric OSA is associated with oxidative stress and inflammation in the airway. OBJECTIVE: The objective of this study is to assess the hypothesis that levels of oxidative stress and inflammatory markers in the exhaled breath condensate (EBC) of children with OSA are higher than those of control subjects. METHODS: Participants were children with OSA and control subjects who underwent overnight polysomnography. Morning levels of hydrogen peroxide (H(2)O(2)) and sum of nitrite and nitrate (NO(x)) in EBC of participants were measured. RESULTS: Twelve subjects with moderate-to-severe OSA (mean age ± standard deviation: 6.3 ± 1.7 years; apnea-hypopnea index--AHI, 13.6 ± 10.1 episodes/h), 22 subjects with mild OSA (6.7 ± 2.1 years; AHI, 2.8 ± 1 episodes/h) and 16 control participants (7.7 ± 2.4 years; AHI, 0.6 ± 0.3 episodes/h) were recruited. Children with moderate-to severe OSA had higher log-transformed H(2)O(2) concentrations in EBC compared to subjects with mild OSA, or to control participants: 0.4 ± 1.1 versus -0.9 ± 1.3 (p = 0.015), or versus -1.2 ± 1.2 (p = 0.003), respectively. AHI and % sleep time with oxygen saturation of hemoglobin <95% were significant predictors of log-transformed H(2)O(2) after adjustment by age and body mass index z score (p < 0.05). No significant differences were demonstrated between the three study groups in terms of EBC NO(x) levels. CONCLUSIONS: Children with moderate-to-severe OSA have increased H(2)O(2) levels in morning EBC, an indirect index of altered redox status in the respiratory tract.


Assuntos
Testes Respiratórios , Mediadores da Inflamação/análise , Estresse Oxidativo/fisiologia , Apneia Obstrutiva do Sono/diagnóstico , Apneia Obstrutiva do Sono/fisiopatologia , Adolescente , Criança , Feminino , Humanos , Peróxido de Hidrogênio/análise , Masculino , Valor Preditivo dos Testes , Valores de Referência , Sistema Respiratório/fisiopatologia
13.
Int J Urol ; 19(6): 565-74, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22324515

RESUMO

OBJECTIVES: Neuropeptides are important signal initiators in advanced prostate cancer, partially acting through activation of nuclear factor kappa B. Central to nuclear factor kappa B regulation is the ubiquitin-proteasome system, pharmacological inhibition of which has been proposed as an anticancer strategy. We investigated the putative role of the proteasome inhibitor bortezomib in neuropeptides signaling effects on prostate cancer cells. METHODS: Human prostate cancer cell lines, LNCaP and PC-3, were used to examine cell proliferation, levels of proapoptotic (caspase-3, Bad) and cell cycle regulatory proteins (p53, p27, p21), as well as total and phosphorylated Akt and p44/42 mitogen-activated protein kinase proteins. Furthermore, 20S proteasome activity, subcellular localization of nuclear factor kappa B and transcription of nuclear factor kappa B target genes, interleukin-8 and vascular endothelial growth factor, were assessed. RESULTS: Neuropeptides (endothelin-1, bombesin) increased cell proliferation, whereas bortezomib decreased proliferation and induced apoptosis, an effect maintained after cotreatment with neuropeptides. Bad, p53, p21 and p27 were downregulated by neuropeptides in PC-3, and these effects were reversed with the addition of bortezomib. Neuropeptides increased proteasomal activity and nuclear factor kappa B levels in PC-3, and these effects were prevented by bortezomib. Interleukin-8 and vascular endothelial growth factor transcripts were induced after neuropeptides treatment, but downregulated by bortezomib. These results coincided with the ability of bortezomib to reduce mitogen-activated protein kinase signaling in both cell lines. CONCLUSIONS: These findings are consistent with bortezomib-mediated abrogation of neuropeptides-induced proliferative and antiapoptotic signaling. Thus, the effect of the drug on the neuropeptides axis needs to be further investigated, as neuropeptide action in prostate cancer might entail involvement of the proteasome.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Bombesina/efeitos dos fármacos , Ácidos Borônicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Endotelina-1/efeitos dos fármacos , Neoplasias da Próstata/patologia , Pirazinas/farmacologia , Bombesina/fisiologia , Bortezomib , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Inibidor de Quinase Dependente de Ciclina p27/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Regulação para Baixo , Endotelina-1/fisiologia , Humanos , Interleucina-8/efeitos dos fármacos , Interleucina-8/genética , Masculino , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , NF-kappa B/efeitos dos fármacos , NF-kappa B/fisiologia , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/fisiologia , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Translocação Genética/efeitos dos fármacos , Proteína Supressora de Tumor p53/efeitos dos fármacos , Proteína Supressora de Tumor p53/fisiologia , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/genética , Proteína de Morte Celular Associada a bcl/efeitos dos fármacos , Proteína de Morte Celular Associada a bcl/fisiologia
14.
J BUON ; 25(2): 1141-1147, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32521918

RESUMO

PURPOSE: Growth factors such as fibroblast growth factor 2 (FGF-2) and hepatocyte growth factor (HGF) appear at high levels in prostate cancer (PC). Abiraterone is an androgen biosynthesis inhibitor which is currently in use as a standard treatment in clinics to impair tumor growth. Development of resistance to anticancer therapies is unfortunately a very common feature of cancer cells that threatens the patient lives. This study aimed to investigate whether FGF-2 and HGF act as a possible resistant mechanism to the abiraterone activity on the androgen synthesis pathway in PC. METHODS: The intracellular levels of 17-OH progesterone and dihydrotestosterone (DHT) were determined by enzyme immunoassays in cell lysates of LNCaP and PC3 PC cells upon co-treatment of cells with abiraterone and FGF-2 or HGF. RESULTS: Abiraterone treatment resulted in significant reduction in the intracellular levels of 17-OH progesterone and DHT in both LnCap and PC3 cells. FGF-2 and HGF were found to decrease the intracellular levels of 17-OH progesterone in both cell lines, whereas HGF alone was found to increase the intracellular levels of DHT only in PC3 cells. However, the simultaneous exposure of cells to abiraterone and FGF-2 or HGF was found to result in an increase in the intracellular levels of DHT, while it did not result in changes in the intracellular levels of 17-OH progesterone. CONCLUSION: These findings suggest that FGF-2 and HGF may act as an escape mechanism, aiding the development of resistance to abiraterone by restoring intra-tumoral androgen synthesis that may contribute to disease progression.


Assuntos
Androstenos/farmacologia , Di-Hidrotestosterona/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Humanos , Masculino , Neoplasias da Próstata/patologia
15.
Am J Crit Care ; 29(1): 15-21, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31968080

RESUMO

BACKGROUND: This study was conducted to investigate whether high-tidal-volume mechanical ventilation is associated with increased lung inflammation compared with low-tidal-volume mechanical ventilation in critically ill patients with no evidence of lung injury. METHODS: In this prospective, single-blind, randomized (1:1), parallel-group study, 18 critically ill patients with normal lungs were randomly assigned to receive mechanical ventilation with a tidal volume of either 6 mL/kg (low tidal volume) or 12 mL/kg (high tidal volume) during the first 4 days in the intensive care unit. RESULTS: At baseline and at 24, 48, and 96 hours, exhaled breath condensate was collected to measure interleukin 1ß, interleukin 10, tumor necrosis factor α, and total nitric oxide metabolites. Interleukin 1ß levels in exhaled breath condensate were significantly increased at 24 hours compared with baseline in the high-tidal-volume group but not in the low-tidal-volume group. The interleukin 1ß increase in the high-tidal-volume group was transient. Exhaled breath condensate levels of interleukin 1ß, interleukin 10, tumor necrosis factor α, and total nitric oxide metabolites did not differ significantly between the high-tidal-volume and low-tidal-volume groups at any time point. CONCLUSION: Short-term mechanical ventilation with a tidal volume of 12 mL/kg may trigger inflammatory responses in the lungs of intensive care unit patients without preexisting lung injury.


Assuntos
Pneumonia/fisiopatologia , Respiração Artificial , Volume de Ventilação Pulmonar/fisiologia , APACHE , Adulto , Idoso , Biomarcadores/análise , Feminino , Grécia , Humanos , Unidades de Terapia Intensiva , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Método Simples-Cego
16.
Helicobacter ; 14(3): 177-9, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19702847

RESUMO

Entire Helicobacter Pylori Neutrophil Activated Protein (HPNAP) and its truncated forms NH(2)-terminal region HPNAP(1-57) and C-terminal region HPNAP(58-144) after cloning into pET29c vector, purification and removal of LPS traces were subjected to human neutrophil activation. Our results revealed that the C-terminal region of HPNAP is indispensable for human neutrophil stimulation and their further adhesion to endothelial cells - a step necessary to H. pylori inflammation - in a ratio equal to that exhibited by the entire protein. In addition, experiments concerning the implication of Arabino-Galactan-Proteins (AGPs) derived from Chios Mastic Gum (CMG), the natural resin of the plant Pistacia lentiscus var. Chia revealed the inhibition of neutrophil activation and therefore their adhesion to endothelial cells, in vitro. Both, the involvement of HPNAP C-terminal region in stimulation-adhesion of neutrophils to endothelial cells as well as the inhibition of this process by AGPs have to be further investigated and may be exploited in a future anti-inflammatory therapy for H. pylori patients.


Assuntos
Proteínas de Bactérias/imunologia , Adesão Celular , Células Endoteliais/fisiologia , Helicobacter pylori/imunologia , Neutrófilos/imunologia , Humanos , Recém-Nascido
17.
Int J Pediatr Otorhinolaryngol ; 113: 218-222, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30173989

RESUMO

OBJECTIVES: In children with snoring, increased production of leukotriene B4 (LTB4) may promote tonsillar hypertrophy and sleep-disordered breathing (SDB) or conversely SDB may enhance LTB4 synthesis. We explored whether: i) high LTB4 serum levels predict tonsillar hypertrophy; and ii) SDB severity correlates with LTB4 serum concentration. METHODS: Normal-weight children with SDB or controls underwent polysomnography and measurement of LTB4 serum concentration. Tonsillar hypertrophy was the main outcome measure and high LTB4 serum level (>75 t h percentile value in controls) was the primary explanatory variable. Odds ratio (OR) and the corresponding 95% confidence intervals (CI) for tonsillar hypertrophy in children with versus without high LTB4 level were calculated. The control subgroup and subgroups of subjects with increasing SDB severity were compared regarding LTB4 concentration by Kruskal-Wallis test. Spearman's correlation co-efficient was applied to assess the association of LTB4 concentration with SDB severity. RESULTS: A total of 104 children with SDB and mean obstructive apnea-hypopnea index-AHI of 4.8 ±â€¯5.3 episodes/h (primary snoring: n = 19; mild SDB: n = 49; moderate/severe SDB: n = 36) and 13 controls (no snoring; AHI: 0.4 ±â€¯0.2 episodes/h) were recruited. The four study subgroups were similar regarding LTB4 serum concentration (P = 0.64). High LTB4 (>170.3 pg/mL) was a significant predictor of tonsillar hypertrophy after adjustment for age and gender (OR 3.0 [1.2-7.2]; P = 0.01). There was no association between AHI or desaturation index and LTB4 serum concentration (r = -0.08; P = 0.37 and r = -0.1; P = 0.30, respectively). CONCLUSION: No association was identified between SDB severity and LTB4 levels, but high LTB4 concentration predicted tonsillar hypertrophy.


Assuntos
Leucotrieno B4/sangue , Tonsila Palatina/patologia , Síndromes da Apneia do Sono/sangue , Adolescente , Criança , Pré-Escolar , Feminino , Humanos , Hipertrofia/patologia , Masculino , Polissonografia/métodos , Índice de Gravidade de Doença , Síndromes da Apneia do Sono/etiologia , Ronco/etiologia
18.
Ann N Y Acad Sci ; 1095: 274-91, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17404040

RESUMO

The objective of this article is to investigate the influence of endothelin-1 (ET-1) on human monocyte Na(+)/H(+) exchanger (NHE) activity and on the atherosclerosis-related monocyte functions. ET-1 caused an increase in pHi and in (22)Na influx of monocytes. A reversal of ET-1 effect on pHi was observed in the presence of the NHE1 inhibitor, cariporide. In addition, the activation of NHE1 by ET-1 was mediated via protein kinase C (PKC), mitogen-activated protein kinase (MAPK), phosphatidylinositol 3-kinase (PI3K), and NADPH oxidase. Also, a link between ET-1 and nitric oxide (NO) was observed. Furthermore, after ET-1 treatment, an increase of the adhesive capacity, the migration ability on laminin and CD36 expression of monocytes, was observed; using cariporide this increase was abolished. Our results showed that ET-1 induces a signaling pathway with the involvement of PKC, MAPK, PI3K, and NADPH oxidase where NHE1 plays a key role. ET-1 also plays a significant role in atherosclerosis-related functions of human monocytes, via NHE1 activation.


Assuntos
Aterosclerose/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Endotelina-1/fisiologia , Monócitos/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Adulto , Humanos , Concentração de Íons de Hidrogênio , Líquido Intracelular/metabolismo , Trocador 1 de Sódio-Hidrogênio
19.
Clin Biochem ; 40(3-4): 248-54, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17196578

RESUMO

OBJECTIVES: The application of a novel assay for the direct measurement of prooxidant-antioxidant balance (PAB) in type II diabetes and the evaluation of antioxidant therapy. DESIGN AND METHODS: The assay is based on 3,3',5,5'-tetramethylbenzidine and its cation, used as a redox indicator participating in two simultaneous reactions. PAB was determined in the sera of healthy volunteers and type II diabetes patients. The results were compared with clinical and biological parameters, protein oxidation markers, as well as the results of antioxidant and prooxidant assays. PAB, after administration of vitamins C and E for 1 day, 1 month and 2 months was also determined. RESULTS: Increased PAB was found in the patients' group and correlated with parameters involved in diabetic complications, protein oxidation markers, antioxidant and prooxidant assays. One day after vitamin administration, a significant shift of PAB towards antioxidants was observed. PAB remained unchanged after 1 month and changed marginally in favor of prooxidants in the second month of the therapy. CONCLUSIONS: These results indicate that the measurement of PAB may be useful to identify and follow-up patients who need antioxidant therapy.


Assuntos
Antioxidantes/análise , Antioxidantes/uso terapêutico , Ácido Ascórbico/uso terapêutico , Técnicas de Laboratório Clínico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Oxidantes/sangue , Vitamina A/uso terapêutico , Adulto , Idoso , Ácido Ascórbico/administração & dosagem , Benzidinas/química , Compostos Cromogênicos/química , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Sensibilidade e Especificidade , Vitamina A/administração & dosagem
20.
Int J Biochem Cell Biol ; 45(11): 2359-68, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23958427

RESUMO

Hypoxia-inducible factors (HIFs) are transcriptional regulators that mediate the cellular response to low oxygen. Although HIF-1 is usually considered as the principal mediator of hypoxic adaptation, several tissues and different cell types express both HIF-1 and HIF-2 isoforms under hypoxia or when treated with hypoxia mimetic chemicals such as cobalt. However, the similarities or differences between HIF-1 and HIF-2, in terms of their tissue- and inducer-specific activation and function, are not adequately characterized. To address this issue, we investigated the effects of true hypoxia and hypoxia mimetics on HIF-1 and HIF-2 induction and specific gene transcriptional activity in two hepatic cancer cell lines, Huh7 and HepG2. Both hypoxia and cobalt caused rapid induction of both HIF-1α and HIF-2α proteins. Hypoxia induced erythropoietin (EPO) expression and secretion in a HIF-2-dependent way. Surprisingly, however, EPO expression was not induced when cells were treated with cobalt. In agreement, both HIF-1- and HIF-2-dependent promoters (of PGK and SOD2 genes, respectively) were activated by hypoxia while cobalt only activated the HIF-1-dependent PGK promoter. Unlike cobalt, other hypoxia mimetics such as DFO and DMOG activated both types of promoters. Furthermore, cobalt impaired the hypoxic stimulation of HIF-2, but not HIF-1, activity and cobalt-induced HIF-2α interacted poorly with USF-2, a HIF-2-specific co-activator. These data show that, despite similar induction of HIF-1α and HIF-2α protein expression, HIF-1 and HIF-2 specific gene activating functions respond differently to different stimuli and suggest the operation of oxygen-independent and gene- or tissue-specific regulatory mechanisms involving additional transcription factors or co-activators.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Cobalto/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Hepáticas/genética , Aminoácidos Dicarboxílicos/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/antagonistas & inibidores , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/genética , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Desferroxamina/farmacologia , Eritropoetina/genética , Eritropoetina/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias Hepáticas/patologia , Fosfoglicerato Quinase/genética , Fosfoglicerato Quinase/metabolismo , Regiões Promotoras Genéticas/genética , Propanolaminas/farmacologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Pirrolidinas/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Superóxido Dismutase/genética , Transcrição Gênica/efeitos dos fármacos , Fatores Estimuladores Upstream/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA