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










Base de dados
Intervalo de ano de publicação
1.
J Cell Physiol ; 232(7): 1754-1760, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27808402

RESUMO

The prevalence of asthma is higher in pre-pubescent and aging males, and in post-pubertal females, strongly indicating that sex steroids (especially estrogen) may be an important modulator in lung disease. We recently demonstrated that airway smooth muscle (ASM) expresses both alpha and beta forms of the estrogen receptor (ERα and ERß) in males and females, and that these receptors regulate intracellular [Ca2+ ] and ASM contractility. Although both ERα and ERß have multiple splice variants, it is unclear if and how the expression of these variants is modulated under conditions such as chronic inflammation/asthma. In order to test the hypothesis that the differential expression of ERα and ERß variants contributes to the pathogenesis of asthma, we profiled the expression of various ERα and ERß genes in asthmatic and inflamed (TNFα- or IL-13-treated) ASM. Gene expression was assessed at both the mRNA and protein levels in asthmatic ASM cells or non-asthmatic cells treated with TNFα (20 ng/ml) or IL-13 (50 ng/ml). We observed marked variation in the expression of ER isoforms in response to inflammatory stimuli, and in non-asthmatic versus asthmatic ASM. Changes in protein levels of ERα and ERß corresponded with the observed differential mRNA patterns. Pharmacological studies implicate cytosolic (p42/44 MAPK and PI3 K) and nuclear (NFκB, STAT6, and AP-1) signaling pathways as putative mechanisms that mediate and/or regulate effects of inflammation on ER expression. We conclude that variations in ASM ER expression profiles occur with inflammation and that ER variants could contribute to estrogen signaling in airway diseases such as asthma. J. Cell. Physiol. 232: 1754-1760, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Inflamação/genética , Pulmão/patologia , Miócitos de Músculo Liso/metabolismo , Receptores de Estrogênio/genética , Asma/genética , Asma/patologia , Sítios de Ligação , Citosol/metabolismo , Humanos , Inflamação/patologia , Miócitos de Músculo Liso/patologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Estrogênio/metabolismo , Transdução de Sinais/genética , Fatores de Transcrição/metabolismo
2.
J Cell Physiol ; 232(5): 1053-1068, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27474898

RESUMO

Long-term exposure to cigarette smoke (CS) triggers airway hyperreactivity and remodeling, effects that involve airway smooth muscle (ASM). We previously showed that CS destabilizes the networked morphology of mitochondria in human ASM by regulating the expression of mitochondrial fission and fusion proteins via multiple signaling mechanisms. Emerging data link regulation of mitochondrial morphology to cellular structure and function. We hypothesized that CS-induced changes in ASM mitochondrial morphology detrimentally affect mitochondrial function, leading to CS effects on contractility and remodeling. Here, ASM cells were exposed to 1% cigarette smoke extract (CSE) for 48 h to alter mitochondrial fission/fusion, or by inhibiting the fission protein Drp1 or the fusion protein Mfn2. Mitochondrial function was assessed via changes in bioenergetics or altered rates of proliferation and apoptosis. Our results indicate that both exposure to CS and inhibition of mitochondrial fission/fusion proteins affect mitochondrial function (i.e., energy metabolism, proliferation, and apoptosis) in ASM cells. In vivo, the airways in mice chronically exposed to CS are thickened and fibrotic, and the expression of proteins involved in mitochondrial function is dramatically altered in the ASM of these mice. We conclude that CS-induced changes in mitochondrial morphology (fission/fusion balance) correlate with mitochondrial function, and thus may control ASM proliferation, which plays a central role in airway health. J. Cell. Physiol. 232: 1053-1068, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Pulmão/patologia , Mitocôndrias/metabolismo , Miócitos de Músculo Liso/metabolismo , Fumar , Trifosfato de Adenosina/biossíntese , Animais , Apoptose , Biomarcadores/metabolismo , Proliferação de Células , Metabolismo Energético , Regulação da Expressão Gênica , Glicólise , Humanos , Potencial da Membrana Mitocondrial , Camundongos Endogâmicos C57BL , Modelos Biológicos , Biogênese de Organelas
3.
Am J Physiol Lung Cell Mol Physiol ; 311(2): L270-9, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27317689

RESUMO

Brain-derived neurotrophic factor (BDNF), a neurotrophin produced by airway smooth muscle (ASM), enhances inflammation effects on airway contractility, supporting the idea that locally produced growth factors influence airway diseases such as asthma. We endeavored to dissect intrinsic mechanisms regulating endogenous, as well as inflammation (TNF-α)-induced BDNF secretion in ASM of nonasthmatic vs. asthmatic humans. We focused on specific Ca(2+) regulation- and inflammation-related signaling cascades and quantified BDNF secretion. We find that TNF-α enhances BDNF release by ASM cells, via several mechanisms relevant to asthma, including transient receptor potential channels TRPC3 and TRPC6 (but not TRPC1), ERK 1/2, PI3K, PLC, and PKC cascades, Rho kinase, and transcription factors cAMP response element binding protein and nuclear factor of activated T cells. Basal BDNF expression and secretion are elevated in asthmatic ASM and increase further with TNF-α exposure, involving many of these regulatory mechanisms. We conclude that airway BDNF secretion is regulated at multiple levels, providing a basis for autocrine effects of BDNF under conditions of inflammation and disease, with potential downstream influences on contractility and remodeling.


Assuntos
Asma/metabolismo , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Miócitos de Músculo Liso/fisiologia , Remodelação das Vias Aéreas , Asma/fisiopatologia , Brônquios/patologia , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Expressão Gênica , Humanos , Contração Muscular , Músculo Liso/patologia , Regiões Promotoras Genéticas , Via Secretória , Transdução de Sinais , Canais de Cátion TRPC/metabolismo
4.
Sci Transl Med ; 7(284): 284ra60, 2015 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-25904744

RESUMO

Airway hyperresponsiveness and inflammation are fundamental hallmarks of allergic asthma that are accompanied by increases in certain polycations, such as eosinophil cationic protein. Levels of these cations in body fluids correlate with asthma severity. We show that polycations and elevated extracellular calcium activate the human recombinant and native calcium-sensing receptor (CaSR), leading to intracellular calcium mobilization, cyclic adenosine monophosphate breakdown, and p38 mitogen-activated protein kinase phosphorylation in airway smooth muscle (ASM) cells. These effects can be prevented by CaSR antagonists, termed calcilytics. Moreover, asthmatic patients and allergen-sensitized mice expressed more CaSR in ASMs than did their healthy counterparts. Indeed, polycations induced hyperreactivity in mouse bronchi, and this effect was prevented by calcilytics and absent in mice with CaSR ablation from ASM. Calcilytics also reduced airway hyperresponsiveness and inflammation in allergen-sensitized mice in vivo. These data show that a functional CaSR is up-regulated in asthmatic ASM and targeted by locally produced polycations to induce hyperresponsiveness and inflammation. Thus, calcilytics may represent effective asthma therapeutics.


Assuntos
Asma/patologia , Asma/fisiopatologia , Hiper-Reatividade Brônquica/metabolismo , Hipersensibilidade/patologia , Receptores de Detecção de Cálcio/antagonistas & inibidores , Alérgenos/química , Animais , Asma/metabolismo , Biópsia , Brônquios/metabolismo , Brônquios/patologia , Líquido da Lavagem Broncoalveolar , Broncoconstrição , Cátions , Células HEK293 , Homeostase , Humanos , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fosforilação , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
Am J Physiol Lung Cell Mol Physiol ; 306(9): L840-54, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24610934

RESUMO

The balance between mitochondrial fission and fusion is crucial for mitochondria to perform its normal cellular functions. We hypothesized that cigarette smoke (CS) disrupts this balance and enhances mitochondrial dysfunction in the airway. In nonasthmatic human airway smooth muscle (ASM) cells, CS extract (CSE) induced mitochondrial fragmentation and damages their networked morphology in a concentration-dependent fashion, via increased expression of mitochondrial fission protein dynamin-related protein 1 (Drp1) and decreased fusion protein mitofusin (Mfn) 2. CSE effects on Drp1 vs. Mfn2 and mitochondrial network morphology involved reactive oxygen species (ROS), activation of extracellular signal-regulated kinase (ERK), phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt), protein kinase C (PKC) and proteasome pathways, as well as transcriptional regulation via factors such as NF-κB and nuclear erythroid 2-related factor 2. Inhibiting Drp1 prevented CSE effects on mitochondrial networks and ROS generation, whereas blocking Mfn2 had the opposite, detrimental effect. In ASM from asmatic patients, mitochondria exhibited substantial morphological defects at baseline and showed increased Drp1 but decreased Mfn2 expression, with exacerbating effects of CSE. Overall, these results highlight the importance of mitochondrial networks and their regulation in the context of cellular changes induced by insults such as inflammation (as in asthma) or CS. Altered mitochondrial fission/fusion proteins have a further potential to influence parameters such as ROS and cell proliferation and apoptosis relevant to airway diseases.


Assuntos
Fragmentação do DNA , DNA Mitocondrial/efeitos dos fármacos , Mitocôndrias Musculares/patologia , Doenças Mitocondriais/etiologia , Músculo Liso/patologia , Fumar/efeitos adversos , Biomarcadores/metabolismo , Western Blotting , Células Cultivadas , Dinaminas , GTP Fosfo-Hidrolases/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Doenças Mitocondriais/metabolismo , Doenças Mitocondriais/patologia , Proteínas Mitocondriais/metabolismo , Músculo Liso/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio
7.
J Mol Cell Cardiol ; 68: 89-97, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24462831

RESUMO

Within human pulmonary artery, neurotrophin growth factors [NTs; e.g. brain-derived neurotrophic factor (BDNF)] and their high-affinity receptors (tropomyosin-related kinase; Trk) and low-affinity receptors p75 neurotrophin receptor (p75NTR) have been reported, but their functional role is incompletely understood. We tested the hypothesis that BDNF is produced by human pulmonary artery endothelial cells (PAECs). In the context of hypoxia as a risk factor for pulmonary hypertension, we examined the effect of hypoxia on BDNF secretion and consequent autocrine effects on pulmonary endothelium. Initial ELISA analysis of circulating BDNF in 30 healthy human volunteers showed that 72 h exposure to high altitude (~11,000 ft, alveolar PO2 = 100 mmHg) results in higher BDNF compared to samples taken at sea level. Separately, in human PAECs exposed for 24h to normoxia vs. hypoxia (1-3% O2), ELISA of extracellular media showed increased BDNF levels. Furthermore, quantitative PCR of PAECs showed 3-fold enhancement of BDNF gene transcription with hypoxia. In PAECs, BDNF induced NO production (measured using an NO-sensitive fluorescent dye DAF2-DA) that was significantly higher under hypoxic conditions, an effect also noted with the TrkB agonist 7,8-DHF. Importantly, hypoxia-induced NO was blunted by neutralization of secreted BDNF using the chimeric TrkB-Fc. Both hypoxia and BDNF increased iNOS (but not eNOS) mRNA expression. In accordance, BDNF enhancement of NO in hypoxia was not blunted by 50 nM L-NAME (eNOS inhibition) but substantially lower with 100 µM L-NAME (eNOS and iNOS inhibition). Hypoxia and BDNF also induced expression of hypoxia inducible factor 1 alpha (HIF-1α), a subunit of the transcription factor HIF-1, and pharmacological inhibition of HIF-1 diminished hypoxia effects on BDNF expression and secretion, and NO production. These results indicate that human PAECs express and secrete BDNF in response to hypoxia via a HIF-1-regulated pathway.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células Endoteliais/metabolismo , Arginase/metabolismo , Fator Neurotrófico Derivado do Encéfalo/sangue , Fator Neurotrófico Derivado do Encéfalo/genética , Hipóxia Celular , Células Cultivadas , Endotélio Vascular/patologia , Expressão Gênica , Humanos , Hipóxia/sangue , Fator 1 Induzível por Hipóxia/metabolismo , Glicoproteínas de Membrana , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Proteínas Quinases/metabolismo , Proteínas Tirosina Quinases , Artéria Pulmonar/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor trkB , Transdução de Sinais
8.
Expert Rev Respir Med ; 7(6): 631-46, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23978003

RESUMO

Mitochondria are autonomous cellular organelles that oversee a variety of functions such as metabolism, energy production, calcium buffering and cell fate determination. Regulation of their morphology and diverse activities beyond energy production are being recognized as playing major roles in cellular health and dysfunction. This review is aimed at summarizing what is known regarding mitochondrial contributions to pathogenesis of lung diseases. Emphasis is given to understanding the importance of structural and functional aspects of mitochondria in both normal cellular function (based on knowledge from other cell types) and in development and modulation of lung diseases such as asthma, chronic obstructive pulmonary disease, cystic fibrosis and cancer. Emerging techniques that allow examination of mitochondria, and potential strategies to target mitochondria in the treatment of lung diseases are also discussed.


Assuntos
Pneumopatias/fisiopatologia , Mitocôndrias/fisiologia , Humanos , Potencial da Membrana Mitocondrial/fisiologia , Mitocôndrias/ultraestrutura , Dinâmica Mitocondrial/fisiologia
9.
Am J Respir Cell Mol Biol ; 49(4): 601-8, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23713409

RESUMO

Inflammation elevates intracellular Ca(2+) ([Ca(2+)]i) concentrations in airway smooth muscle (ASM). Store-operated Ca(2+) entry (SOCE) is an important source of [Ca(2+)]i mediated by stromal interaction molecule-1 (STIM1), a sarcoplasmic reticulum (SR) protein. In transducing SR Ca(2+) depletion, STIM1 aggregates to form puncta, thereby activating SOCE via interactions with a Ca(2+) release-activated Ca(2+) channel protein (Orai1) in the plasma membrane. We hypothesized that STIM1 aggregation is enhanced by inflammatory cytokines, thereby augmenting SOCE in human ASM cells. We used real-time fluorescence microscopic imaging to assess the dynamics of STIM1 aggregation and SOCE after exposure to TNF-α or IL-13 in ASM cells overexpressing yellow fluorescent protein-tagged wild-type STIM1 (WT-STIM1) and STIM1 mutants lacking the Ca(2+)-sensing EF-hand (STIM1-D76A), or lacking the cytoplasmic membrane binding site (STIM1ΔK). STIM1 aggregation was analyzed by monitoring puncta size during the SR Ca(2+) depletion induced by cyclopiazonic acid (CPA). We found that puncta size was increased in cells expressing WT-STIM1 after CPA. However, STIM1-D76A constitutively formed puncta, whereas STIM1ΔK failed to form puncta. Furthermore, cytokines increased basal WT-STIM1 puncta size, and the SOCE triggered by SR Ca(2+) depletion was increased in cells expressing WT-STIM1 or STIM1-D76A. Meanwhile, SOCE in cells expressing STIM1ΔK and STIM1 short, interfering RNA (siRNA) was decreased. Similarly, in cells overexpressing STIM1, the siRNA knockdown of Orai1 blunted SOCE. However, exposure to cytokines increased SOCE in all cells, increased basal [Ca(2+)]i, and decreased SR Ca(2+) content. These data suggest that cytokines induce a constitutive increase in STIM1 aggregation that contributes to enhanced SOCE in human ASM after inflammation. Such effects of inflammation on STIM1 aggregations may contribute to airway hyperresponsiveness.


Assuntos
Cálcio/metabolismo , Inflamação/metabolismo , Interleucina-13/metabolismo , Proteínas de Membrana/metabolismo , Músculo Liso/metabolismo , Proteínas de Neoplasias/metabolismo , Sistema Respiratório/embriologia , Fator de Necrose Tumoral alfa/metabolismo , Sítios de Ligação , Canais de Cálcio/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Humanos , Miócitos de Músculo Liso/metabolismo , Proteína ORAI1 , Retículo Sarcoplasmático/metabolismo , Molécula 1 de Interação Estromal
10.
Am J Respir Cell Mol Biol ; 48(4): 431-8, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23258230

RESUMO

Enhanced airway smooth muscle (ASM) contractility contributes to increased resistance to airflow in diseases such as bronchitis and asthma that occur in passive smokers exposed to secondhand smoke. Little information exists on the cellular mechanisms underlying such airway hyperreactivity. Sputum samples of patients with chronic sinusitis, bronchitis, and asthma show increased concentrations of growth factors called neurotrophins, including brain-derived growth factor (BDNF), but their physiological significance remains unknown. In human ASM, we tested the hypothesis that BDNF contributes to increased contractility with cigarette smoke exposure. The exposure of ASM to 1% or 2% cigarette smoke extract (CSE) for 24 hours increased intracellular calcium ([Ca(2+)](i)) responses to histamine, and further potentiated the enhancing effects of a range of BDNF concentrations on such histamine responses. CSE exposure increased the expression of the both high-affinity and low-affinity neurotrophin receptors tropomyosin-related kinase (Trk)-B and p75 pan-neurotrophin receptor, respectively. Quantitative ELISA showed that CSE increased BDNF secretion by human ASM cells. BDNF small interfering (si)RNA and/or the chelation of extracellular BDNF, using TrkB-fragment crystallizable, blunted the effects of CSE on [Ca(2+)](i) responses as well as the CSE enhancement of cell proliferation, whereas TrkB siRNA blunted the effects of CSE on ASM contractility. These data suggest that cigarette smoke is a potent inducer of BDNF and TrkB expression and signaling in ASM, which then contribute to cigarette smoke-induced airway hyperresponsiveness.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Hiper-Reatividade Brônquica/metabolismo , Músculo Liso/metabolismo , Fumar/efeitos adversos , Poluição por Fumaça de Tabaco/efeitos adversos , Hiper-Reatividade Brônquica/induzido quimicamente , Hiper-Reatividade Brônquica/patologia , Cálcio/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Histamina/metabolismo , Humanos , Masculino , Contração Muscular/efeitos dos fármacos , Músculo Liso/patologia , Proteínas do Tecido Nervoso/biossíntese , Receptor trkB/biossíntese , Receptores de Fator de Crescimento Neural/biossíntese , Fumar/metabolismo , Fumar/patologia
11.
PLoS One ; 7(8): e44343, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22952960

RESUMO

Neurotrophins (NTs), which play an integral role in neuronal development and function, have been found in non-neuronal tissue (including lung), but their role is still under investigation. Recent reports show that NTs such as brain-derived neurotrophic factor (BDNF) as well as NT receptors are expressed in human airway smooth muscle (ASM). However, their function is still under investigation. We hypothesized that NTs regulate ASM intracellular Ca(2+) ([Ca(2+)](i)) by altered expression of Ca(2+) regulatory proteins. Human ASM cells isolated from lung samples incidental to patient surgery were incubated for 24 h (overnight) in medium (control) or 1 nM BDNF in the presence vs. absence of inhibitors of signaling cascades (MAP kinases; PI3/Akt; NFκB). Measurement of [Ca(2+)](i) responses to acetylcholine (ACh) and histamine using the Ca(2+) indicator fluo-4 showed significantly greater responses following BDNF exposure: effects that were blunted by pathway inhibitors. Western analysis of whole cell lysates showed significantly higher expression of CD38, Orai1, STIM1, IP(3) and RyR receptors, and SERCA following BDNF exposure, effects inhibited by inhibitors of the above cascades. The functional significance of BDNF effects were verified by siRNA or pharmacological inhibition of proteins that were altered by this NT. Overall, these data demonstrate that NTs activate signaling pathways in human ASM that lead to enhanced [Ca(2+)](i) responses via increased regulatory protein expression, thus enhancing airway contractility.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/farmacologia , Cálcio/metabolismo , Músculo Liso/metabolismo , Sistema Respiratório/metabolismo , Flavonas/farmacologia , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Sistema Respiratório/citologia , Retículo Sarcoplasmático/efeitos dos fármacos , Retículo Sarcoplasmático/metabolismo , Transdução de Sinais/efeitos dos fármacos
12.
Am J Physiol Lung Cell Mol Physiol ; 303(8): L711-9, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22923637

RESUMO

Maintenance of blood oxygen saturation dictates supplemental oxygen administration to premature infants, but hyperoxia predisposes survivors to respiratory diseases such as asthma. Although much research has focused on oxygen effects on alveoli in the setting of bronchopulmonary dysplasia, the mechanisms by which oxygen affects airway structure or function relevant to asthma are still under investigation. We used isolated human fetal airway smooth muscle (fASM) cells from 18-20 postconceptual age lungs (canalicular stage) to examine oxygen effects on intracellular Ca(2+) ([Ca(2+)](i)) and cellular proliferation. fASM cells expressed substantial smooth muscle actin and myosin and several Ca(2+) regulatory proteins but not fibroblast or epithelial markers, profiles qualitatively comparable to adult human ASM. Fluorescence Ca(2+) imaging showed robust [Ca(2+)](i) responses to 1 µM acetylcholine (ACh) and 10 µM histamine (albeit smaller and slower than adult ASM), partly sensitive to zero extracellular Ca(2+). Compared with adult, fASM showed greater baseline proliferation. Based on this validation, we assessed fASM responses to 10% hypoxia through 90% hyperoxia and found enhanced proliferation at <60% oxygen but increased apoptosis at >60%, effects accompanied by appropriate changes in proliferative vs. apoptotic markers and enhanced mitochondrial fission at >60% oxygen. [Ca(2+)](i) responses to ACh were enhanced for <60% but blunted at >60% oxygen. These results suggest that hyperoxia has dose-dependent effects on structure and function of developing ASM, which could have consequences for airway diseases of childhood. Thus detrimental effects on ASM should be an additional consideration in assessing risks of supplemental oxygen in prematurity.


Assuntos
Hiperóxia/metabolismo , Hipóxia/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Oxigênio/efeitos adversos , Traqueia/metabolismo , Adulto , Asma/epidemiologia , Asma/metabolismo , Asma/patologia , Cálcio/metabolismo , Proliferação de Células , Células Cultivadas , Feto/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Hiperóxia/epidemiologia , Hiperóxia/patologia , Hipóxia/epidemiologia , Hipóxia/patologia , Recém-Nascido , Recém-Nascido Prematuro , Mitocôndrias/metabolismo , Miócitos de Músculo Liso/citologia , Oxigênio/administração & dosagem , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Fatores de Risco , Traqueia/citologia , Traqueia/embriologia
13.
Am J Physiol Lung Cell Mol Physiol ; 303(8): L669-81, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22923642

RESUMO

Caveolae are flask-shaped plasma membrane invaginations expressing the scaffolding caveolin proteins. Although caveolins have been found in endothelium and epithelium (where they regulate nitric oxide synthase activity), their role in smooth muscle is still under investigation. We and others have previously shown that caveolae of human airway smooth muscle (ASM), which express caveolin-1, contain Ca(2+) and force regulatory proteins and are involved in mediating the effects of inflammatory cytokines such as TNF-α on intracellular Ca(2+) concentration responses to agonist. Accordingly, we tested the hypothesis that in vivo, absence of caveolin-1 leads to reduced airway hyperresponsiveness, using a knockout (KO) (Cav1 KO) mouse and an ovalbumin-sensitized/challenged (OVA) model of allergic airway hyperresponsiveness. Surprisingly, airway responsiveness to methacholine, tested by use of a FlexiVent system, was increased in Cav1 KO control (CTL) as well as KO OVA mice, which could not be explained by a blunted immune response to OVA. In ASM of wild-type (WT) OVA mice, expression of caveolin-1, the caveolar adapter proteins cavins 1-3, and caveolae-associated Ca(2+) and force regulatory proteins such as Orai1 and RhoA were all increased, effects absent in Cav1 KO CTL and OVA mice. However, as with WT OVA, both CTL and OVA Cav1 KO airways showed signs of enhanced remodeling, with high expression of proliferation markers and increased collagen. Separately, epithelial cells from airways of all three groups displayed lower endothelial but higher inducible nitric oxide synthase and arginase expression. Arginase activity was also increased in these three groups, and the inhibitor nor-NOHA (N-omega-nor-l-arginine) enhanced sensitivity of isolated tracheal rings to ACh, especially in Cav1 KO mice. On the basis of these data disproving our original hypothesis, we conclude that caveolin-1 has complex effects on ASM vs. epithelium, resulting in airway hyperreactivity in vivo mediated by altered airway remodeling and bronchodilation.


Assuntos
Hiper-Reatividade Brônquica/imunologia , Caveolina 1/genética , Caveolina 1/imunologia , Animais , Hiper-Reatividade Brônquica/patologia , Broncoconstrição/efeitos dos fármacos , Broncoconstrição/imunologia , Broncoconstritores/farmacologia , Cálcio/imunologia , Modelos Animais de Doenças , Cloreto de Metacolina/farmacologia , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Músculo Liso/imunologia , Ovalbumina/imunologia , Ovalbumina/farmacologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/patologia
14.
J Cell Mol Med ; 16(4): 812-23, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21651720

RESUMO

Airway diseases such as asthma involve increased airway smooth muscle (ASM) contractility and remodelling via enhanced proliferation. Neurotrophins (NTs) such as brain-derived neurotrophic factor (BDNF), well-known in the nervous system, can regulate Ca(2+) signalling, and interact with cytokines in contributing to airway hyperreactivity. In this study, we determined whether and how BDNF regulates human ASM cell proliferation in the presence of inflammation, thus testing its potential role in airway remodelling. Cells were treated with 10 nM BDNF, 25 ng/ml tumour necrosis factor (TNF-α) or interleukin-13 (IL-13), or 10 ng/ml platelet-derived growth factor (PDGF). Proliferation was measured using CyQuant dye, with immunoblotting of cell cycle proteins predicted to change with proliferation. Forty-eight hours of BDNF enhanced ASM proliferation to ≈ 50% of that by PDGF or cytokines. Transfection with small interfering RNAs (siRNAs) targeting high-affinity tropomyosin-related kinase B receptor abolished BDNF effects on proliferation, whereas low-affinity 75 kD neurotrophin receptor (p75NTR) siRNA had no effect. Systematic pharmacologic inhibition of different components of ERK1/2 and PI3K/Akt1 pathways blunted BDNF or TNF-α-induced proliferation. BDNF also induced IκB phosphorylation and nuclear translocation of p50 and p65 NF-κB subunits, with electron mobility shift assay confirmation of NF-κB binding to consensus DNA sequence. These results demonstrate that NTs such as BDNF can enhance human ASM cell proliferation by activating proliferation-specific signalling pathways and a versatile transcription factor such as NF-κB, which are common to cytokines and growth factors involved in asthma.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/farmacologia , Brônquios/efeitos dos fármacos , Proliferação de Células , Músculo Liso/efeitos dos fármacos , Western Blotting , Brônquios/citologia , Células Cultivadas , Ensaio de Desvio de Mobilidade Eletroforética , Citometria de Fluxo , Humanos , Interleucina-13/farmacologia , Músculo Liso/citologia , Fosforilação , Fator de Crescimento Derivado de Plaquetas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia
15.
Eur J Neurosci ; 27(1): 31-42, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18184313

RESUMO

Exposure to drugs of abuse activates gene expression and protein synthesis that result in long-lasting adaptations in striatal signaling. Therefore, identification of the transcription factors that couple drug exposure to gene expression is of particular importance. Members of the nuclear factor of activated T-cells (NFATc) family of transcription factors have recently been implicated in shaping neuronal function throughout the rodent nervous system. Here we demonstrate that regulation of NFAT-mediated gene expression may also be a factor in drug-induced changes to striatal functioning. In cultured rat striatal neurons, stimulation of D1 dopamine receptors induces NFAT-dependent transcription through activation of L-type calcium channels. Additionally, the genes encoding inositol-1,4,5-trisphosphate receptor type 1 and glutamate receptor subunit 2 are regulated by striatal NFATc4 activity. Consistent with these in-vitro data, repeated exposure to cocaine triggers striatal NFATc4 nuclear translocation and the up-regulation of inositol-1,4,5-trisphosphate receptor type 1 and glutamate receptor subunit 2 gene expression in vivo, suggesting that cocaine-induced increases in gene expression may be partially mediated through activation of NFAT-dependent transcription. Collectively, these findings reveal a novel molecular pathway that may contribute to the enduring modifications in striatal functioning that occur following the administration of drugs of abuse.


Assuntos
Corpo Estriado/metabolismo , Expressão Gênica/fisiologia , Fatores de Transcrição NFATC/metabolismo , Receptores de Dopamina D1/fisiologia , Animais , Animais Recém-Nascidos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células Cultivadas , Imunoprecipitação da Cromatina/métodos , Cocaína/farmacologia , Corpo Estriado/efeitos dos fármacos , Inibidores da Captação de Dopamina/farmacologia , Ensaio de Desvio de Mobilidade Eletroforética/métodos , Agonistas de Aminoácidos Excitatórios/farmacologia , Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Receptores de AMPA/metabolismo , Transfecção/métodos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia
16.
J Appl Physiol (1985) ; 104(3): 787-94, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18187618

RESUMO

During early postnatal development in rat diaphragm muscle (Diam), significant fiber growth and transitions in myosin heavy chain (MHC) isoform expression occur. Similar to other skeletal muscles, Diam fibers are multinucleated, and each myonucleus regulates the gene products within a finite volume: the myonuclear domain (MND). We hypothesized that postnatal changes in fiber cross-sectional area (CSA) are associated with increased number of myonuclei so that the MND size is maintained. The Diam was removed at postnatal days 14 (P-14) and 28 (P-28). MHC isoform expression was determined by SDS-PAGE. Fiber CSA, myonuclear number, and MND size were measured using confocal microscopy. By P-14, significant coexpression of MHC isoforms was present with no fiber displaying singular expression of MHCNeo. By P-28, singular expression was predominant. MND size was not different across fiber types at P-14. Significant fiber growth was evident by P-28 at all fiber types (fiber CSA increased by 61, 93, and 147% at fibers expressing MHCSlow, MHC2A, and MHC2X, respectively). The number of myonuclei per unit of fiber length was similar across fibers at P-14, but it was greater at fibers expressing MHC2X at P-28. The total number of myonuclei per fiber also increased between P-14 and P-28 at all fiber types. Accordingly, MND size increased significantly by P-28 at all fiber types, and it became larger at fibers expressing MHC2X compared with fibers expressing MHCSlow or MHC2A. These results suggest that MND size is not maintained during the considerable fiber growth associated with postnatal development of the Diam.


Assuntos
Núcleo Celular/ultraestrutura , Diafragma/crescimento & desenvolvimento , Desenvolvimento Muscular , Fibras Musculares Esqueléticas/ultraestrutura , Fatores Etários , Animais , Peso Corporal , Crescimento Celular , Núcleo Celular/metabolismo , Tamanho do Núcleo Celular , Diafragma/citologia , Diafragma/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Isoformas de Proteínas/metabolismo , Ratos , Ratos Sprague-Dawley
17.
J Appl Physiol (1985) ; 100(5): 1617-22, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16410375

RESUMO

Denervation (DNV) of rat diaphragm muscle (DIAm) leads to selective atrophy of type IIx and IIb fibers, whereas the cross-sectional area of type I and IIa fibers remains unchanged or slightly hypertrophied. DIAm DNV also increases satellite cell mitotic activity and myonuclear apoptosis. Similar to other skeletal muscles, DIAm fibers are multinucleated, and each myonucleus regulates the gene products in a finite fiber volume, i.e., myonuclear domain (MND). MND size varies across DIAm fiber types in rank order, I < IIa < IIx < IIb [fiber type based on myosin heavy chain isoform expression]. We hypothesized that, after DNV, the total number of myonuclei per fiber does not change and, accordingly, that MND changes proportionately to the change in fiber size regardless of fiber type. Adult rats underwent unilateral (right side) DIAm DNV, and after 2 wk single fibers were dissected. Fiber cross-sectional area, myonuclear number, and MND were measured by confocal microscopy, and these values in DNV DIAm were compared with those obtained in controls. After DNV, type I fibers hypertrophied, type IIa fiber size was unchanged, and type IIx and IIb fibers atrophied compared with control. The total number of myonuclei per fiber was not affected by DNV. Accordingly, after DNV, type I fiber MND increased by 25%, whereas it decreased in type IIx and IIb fibers by 50 and 70%, respectively. These results suggest that MND is not maintained after DNV-induced DIAm fiber hypertrophy or atrophy. These results are interpreted with respect to consequent effects of DNV on myonuclear transcriptional activity and protein turnover.


Assuntos
Núcleo Celular/fisiologia , Diafragma/inervação , Diafragma/fisiopatologia , Denervação Muscular , Fibras Musculares Esqueléticas/patologia , Animais , Apoptose/fisiologia , Peso Corporal/fisiologia , Núcleo Celular/ultraestrutura , Diafragma/patologia , Regulação da Expressão Gênica , Hipertrofia/patologia , Hipertrofia/fisiopatologia , Masculino , Microscopia Confocal , Mitose/fisiologia , Fibras Musculares Esqueléticas/química , Fibras Musculares Esqueléticas/ultraestrutura , Atrofia Muscular/etiologia , Atrofia Muscular/patologia , Atrofia Muscular/fisiopatologia , Cadeias Pesadas de Miosina/análise , Cadeias Pesadas de Miosina/genética , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Músculos Respiratórios/inervação , Músculos Respiratórios/patologia , Músculos Respiratórios/fisiopatologia , Transcrição Gênica/fisiologia
18.
Hum Mol Genet ; 12(21): 2777-88, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12966035

RESUMO

Caveolins are structural protein components of caveolar membrane domains. Caveolin-3, a muscle-specific member of the caveolin family, is expressed in skeletal muscle tissue and in the heart. The multiple roles that caveolin-3 plays in cellular physiology are becoming more apparent. We have shown that lack of caveolin-3 expression in skeletal muscle resembles limb-girdle muscular dystrophy-1C. In contrast, we have demonstrated that overexpression of caveolin-3 in skeletal muscle tissue promotes defects similar to those seen in Duchenne muscular dystrophy (DMD). Thus, a tight regulation of caveolin-3 expression is fundamental for normal muscle functions. Since caveolin-3 is also endogenously expressed in cardiac myocytes, and cardiomyopathies are observed in DMD patients, we looked at the effects of overexpression of caveolin-3 on cardiac structure and function by characterizing caveolin-3 transgenic mice. Our results indicate that overexpression of caveolin-3 causes severe cardiac tissue degeneration, fibrosis and a reduction in cardiac functions. We also show that dystrophin and its associated glycoproteins are down-regulated in caveolin-3 transgenic heart. In addition, we demonstrate that the activity of nitric oxide synthase (NOS) is down-regulated by high levels of caveolin-3 in the heart. Taken together, these results indicate that overexpression of caveolin-3 is sufficient to induce severe cardiomyopathy. In addition, these findings suggest that caveolin-3 transgenic mice may represent a valid mouse model for studying the molecular mechanisms underlying cardiomyopathies associated with Duchenne muscular dystrophy.


Assuntos
Cavéolas/metabolismo , Caveolinas/metabolismo , Distrofias Musculares/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Miocárdio/metabolismo , Animais , Caveolina 1 , Caveolina 3 , Caveolinas/genética , Regulação para Baixo/genética , Regulação para Baixo/fisiologia , Distrofina/genética , Distrofina/metabolismo , Eletrocardiografia , Humanos , Camundongos , Camundongos Transgênicos , Distrofias Musculares/genética , Distrofia Muscular de Duchenne/genética , Miócitos Cardíacos/metabolismo , Óxido Nítrico Sintase/metabolismo
19.
Curr Biol ; 13(11): 899-910, 2003 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-12781128

RESUMO

BACKGROUND: The ubiquitin proteasome system (UPS) mediates regulated protein degradation and provides a mechanism for closely controlling protein abundance in spatially restricted domains within cells. We hypothesized that the UPS may acutely determine the local concentration of key regulatory proteins at neuronal synapses as a means for locally modulating synaptic efficacy and the strength of neurotransmission communication. RESULTS: We investigated this hypothesis at the Drosophila neuromuscular synapse by using an array of genetic and pharmacological tools. This study demonstrates that UPS components are present in presynaptic boutons and that the UPS functions locally in the presynaptic compartment to rapidly eliminate a conditional transgenic reporter of proteasome activity. We assayed a panel of synaptic proteins to determine whether the UPS acutely regulates the local abundance of native synaptic targets. Both acute pharmacological inhibition of the proteasome (<1 hr) and targeted genetic perturbation of proteasome function in the presynaptic neuron cause the specific accumulation of the essential synaptic vesicle-priming protein DUNC-13. Most importantly, acute pharmacological inhibition of the proteasome (<1 hr) causes a rapid strengthening of neurotransmission (an approximately 50% increase in evoked amplitude) because of increased presynaptic efficacy. The proteasome-dependent regulation of presynaptic protein abundance, both of the exogenous reporter and native DUNC-13, and the modulation of presynaptic neurotransmitter release occur on an intermediate, rapid (tens of minutes) timescale. CONCLUSIONS: Taken together, these studies demonstrate that the UPS functions locally within synaptic boutons to acutely control levels of presynaptic protein and that the rate of UPS-dependent protein degradation is a primary determinant of neurotransmission strength.


Assuntos
Cisteína Endopeptidases/metabolismo , Drosophila/metabolismo , Complexos Multienzimáticos/metabolismo , Neurotransmissores/metabolismo , Terminações Pré-Sinápticas/metabolismo , Sinapses/metabolismo , Ubiquitina/metabolismo , Animais , Animais Geneticamente Modificados , Western Blotting , Primers do DNA , Eletrofisiologia , Imuno-Histoquímica , Larva/metabolismo , Microscopia Confocal , Complexos Multienzimáticos/antagonistas & inibidores , Terminações Pré-Sinápticas/química , Complexo de Endopeptidases do Proteassoma
20.
J Neurobiol ; 54(3): 417-38, 2003 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-12532395

RESUMO

UNC-13 is a highly conserved plasma membrane-associated synaptic protein implicated in the regulation of neurotransmitter release through the direct modulation of the SNARE exocytosis complex. Previously, we characterized the Drosophila homologue (DUNC-13) and showed it to be essential for neurotransmitter release immediately upstream of vesicular fusion ("priming") at the neuromuscular junction (NMJ). Here, we show that the abundance of DUNC-13 in NMJ synaptic boutons is regulated downstream of GalphaS and Galphaq pathways, which have inhibitory and facilitatory roles, respectively. Both cAMP modulation and PKA function are required for DUNC-13 synaptic up-regulation, suggesting that the cAMP pathway enhances synaptic efficacy via DUNC-13. Similarly, PLC function and DAG modulation also regulate the synaptic levels of DUNC-13, through a mechanism that appears independent of PKC. Our results suggest that proteasome-mediated protein degradation is the primary mechanism regulating DUNC-13 levels at the synapse. Both PLC- and PKA-mediated pathways appear to regulate synaptic levels of DUNC-13 through controlling the rate of proteasome-dependent DUNC-13 degradation. We conclude that the functional abundance of DUNC-13 at the synapse, a key determinant of synaptic vesicle priming and neurotransmitter release probability, is primarily regulated by the rate of protein degradation, rather than translocation or transport, convergently controlled via both cAMP and DAG signal transduction pathways.


Assuntos
Acetilcisteína/análogos & derivados , Proteínas de Caenorhabditis elegans , Carbazóis , Cisteína Endopeptidases/metabolismo , Drosophila/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Proteínas de Helminto/metabolismo , Complexos Multienzimáticos/metabolismo , Sinapses/metabolismo , Acetilcisteína/farmacologia , Animais , Anticorpos , Western Blotting , Proteínas de Transporte , Colforsina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Drosophila/anatomia & histologia , Proteínas de Drosophila/efeitos dos fármacos , Proteínas de Drosophila/metabolismo , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Estrenos/farmacologia , Proteínas de Ligação ao GTP/antagonistas & inibidores , Proteínas de Ligação ao GTP/classificação , Peroxidase do Rábano Silvestre/metabolismo , Imuno-Histoquímica , Indóis/farmacologia , Proteínas de Insetos/metabolismo , Maleimidas/farmacologia , Microscopia Confocal/instrumentação , Microscopia Confocal/métodos , Mutação , Neomicina/farmacologia , Junção Neuromuscular/efeitos dos fármacos , Junção Neuromuscular/metabolismo , Oligopeptídeos/farmacologia , Ésteres de Forbol/farmacologia , Éteres Fosfolipídicos/farmacologia , Terminações Pré-Sinápticas/classificação , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/metabolismo , Complexo de Endopeptidases do Proteassoma , Pirróis/farmacologia , Pirrolidinonas/farmacologia , Proteínas Supressoras de Tumor/metabolismo , Fosfolipases Tipo C/efeitos dos fármacos , Fosfolipases Tipo C/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...