Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 277
Filtrar
1.
Int J Mol Sci ; 24(14)2023 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-37511176

RESUMO

In rats, acute normobaric hypoxia depressed left ventricular (LV) inotropic function. After 24 h of hypoxic exposure, a slight recovery of LV function occurred. We speculated that prolonged hypoxia (72 h) would induce acclimatization and, hence, recovery of LV function. Moreover, we investigated biomarkers of nitrosative stress and apoptosis as possible causes of hypoxic LV depression. To elucidate the role of hypoxic sympathetic activation, we studied whether adrenergic blockade would further deteriorate the general state of the animals and their cardiac function. Ninety-four rats were exposed over 72 h either to normal room air (N) or to normobaric hypoxia (H). The rodents received infusion (0.1 mL/h) with 0.9% NaCl or with different adrenergic blockers. Despite clear signs of acclimatization to hypoxia, the LV depression continued persistently after 72 h of hypoxia. Immunohistochemical analyses revealed significant increases in markers of nitrosative stress, adenosine triphosphate deficiency and apoptosis in the myocardium, which could provide a possible explanation for the absence of LV function recovery. Adrenergic blockade had a slightly deteriorative effect on the hypoxic LV function compared to the hypoxic group with maintained sympathetic efficacy. These findings show that hypoxic sympathetic activation compensates, at least partially, for the compromised function in hypoxic conditions, therefore emphasizing its importance for hypoxia adaptation.


Assuntos
Hipóxia , Miocárdio , Ratos , Animais , Função Ventricular Esquerda , Aclimatação , Adrenérgicos/farmacologia
2.
Am J Physiol Lung Cell Mol Physiol ; 324(5): L639-L651, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36648147

RESUMO

Idiopathic pulmonary fibrosis is increasingly associated with nerve-driven processes and endogenous innate immune ligands such as mitochondrial DNA (mtDNA). Interestingly, a connection between these entities has not been explored. Here, we report that noradrenaline (NA) derived from the lung's adrenergic nerve supply drives α-smooth muscle actin (αSMA)-expressing fibroblast accumulation via mechanisms involving α1 adrenoreceptors and mtDNA. Using the bleomycin model, we compared ablation of the lung's adrenergic nerve supply with surgical adrenal resection and found that NA derived from local but not adrenal sources contributes to experimentally induced lung fibrosis and the emergence of an αSMA+ve fibroblast population expressing adrenoreceptor α-1D (ADRA1D). Therapeutic delivery of the α1 adrenoreceptor antagonist terazosin reversed these changes and suppressed extracellular mtDNA accumulation. Cultured normal human lung fibroblasts displayed α1 adrenoreceptors and in response to costimulation with TGFß1 and NA adopted ACTA2 expression and extracellular mtDNA release. These findings were opposed by terazosin. Evaluation of a previously studied IPF cohort revealed that patients prescribed α1 adrenoreceptor antagonists for nonpulmonary indications demonstrated improved survival and reduced concentrations of plasma mtDNA. Our observations link nerve-derived NA, α1 adrenoreceptors, extracellular mtDNA, and lung fibrogenesis in mouse models, cultured cells, and humans with IPF. Further study of this neuroinnate connection may yield new avenues for investigation in the clinical and basic science realms.


Assuntos
DNA Mitocondrial , Fibrose Pulmonar Idiopática , Camundongos , Animais , Humanos , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Fibrose Pulmonar Idiopática/metabolismo , Pulmão/metabolismo , Transdução de Sinais , Fibroblastos/metabolismo , Bleomicina/farmacologia , Adrenérgicos/metabolismo , Adrenérgicos/farmacologia
3.
BMC Neurosci ; 24(1): 2, 2023 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-36631757

RESUMO

BACKGROUND: The head-twitch response (HTR) in mice is considered a behavioral model for hallucinogens and serotonin 5-HT2A receptor function, as well as Tourette syndrome in humans. It is mediated by 5-HT2A receptor agonists such as ( ±)- 2,5-dimethoxy-4-iodoamphetamine (DOI) in the prefrontal cortex (PFC). The 5-HT2A antagonist EMD 281014, can prevent both DOI-induced HTR during ageing and c-fos expression in different regions of PFC. Moreover, the nonselective monoamine releaser methamphetamine (MA) suppressed DOI-induced HTR through ageing via concomitant activation of inhibitory 5-HT1A receptors, but enhanced DOI-evoked c-fos expression. d-Fenfluramine is a selective 5-HT releaser and induces HTR in mice, whereas MA does not. Currently, we investigated whether EMD 281014 or MA would alter: (1) d-fenfluramine-induced HTR frequency in 20-, 30- and 60-day old mice, (2) d-fenfluramine-evoked c-fos expression in PFC, and (3) whether blockade of inhibitory serotonergic 5-HT1A- or adrenergic ɑ2-receptors would prevent suppressive effect of MA on d-fenfluramine-induced HTR. RESULTS: EMD 281014 (0.001-0.05 mg/kg) or MA (0.1-5 mg/kg) blocked d-fenfluramine-induced HTR dose-dependently during ageing. The 5-HT1A antagonist WAY 100635 countered the inhibitory effect of MA on d-fenfluramine-induced HTR in 30-day old mice, whereas the adrenergic ɑ2 antagonist RS 79948 reversed MA's inhibitory effect in both 20- and 30- day old mice. d-Fenfluramine significantly increased c-fos expressions in PFC regions. MA (1 mg/kg) pretreatment significantly increased d-fenfluramine-evoked c-fos expression in different regions of PFC. EMD 281014 (0.05 mg/kg) failed to prevent d-fenfluramine-induced c-fos expression, but significantly increased it in one PFC region (PrL at - 2.68 mm). CONCLUSION: EMD 281014 suppressed d-fenfluramine-induced HTR but failed to prevent d-fenfluramine-evoked c-fos expression which suggest involvement of additional serotonergic receptors in the mediation of evoked c-fos. The suppressive effect of MA on d-fenfluramine-evoked HTR is due to well-recognized functional interactions between stimulatory 5-HT2A- and the inhibitory 5-HT1A- and ɑ2-receptors. MA-evoked increases in c-fos expression in PFC regions are due to the activation of diverse monoaminergic receptors through increased synaptic concentrations of 5-HT, NE and/or DA, which may also account for the additive effect of MA on d-fenfluramine-evoked changes in c-fos expression. Our findings suggest potential drug receptor functional interaction during development when used in combination.


Assuntos
Fenfluramina , Metanfetamina , Córtex Pré-Frontal , Proteínas Proto-Oncogênicas c-fos , Animais , Humanos , Camundongos , Adrenérgicos/metabolismo , Adrenérgicos/farmacologia , Envelhecimento/metabolismo , Fenfluramina/metabolismo , Fenfluramina/farmacologia , Metanfetamina/metabolismo , Metanfetamina/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Receptor 5-HT2A de Serotonina/efeitos dos fármacos , Receptor 5-HT2A de Serotonina/metabolismo , Serotonina/metabolismo , Proteínas Proto-Oncogênicas c-fos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo
4.
Nutrients ; 14(23)2022 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-36500973

RESUMO

Isoflavone-rich legumes, including soy, are used for food production, as dietary supplements and in traditional medicine. Soy consumption correlates negatively with benign prostatic hyperplasia (BPH) and voiding symptoms. However, isoflavone effects on the prostate are hardly known. Here, we examined the effects on human prostate smooth muscle contractions and stromal cell growth, which are driving factors of voiding symptoms in BPH. Smooth muscle contractions were induced in prostate tissues from radical prostatectomy. Growth-related functions were studied in cultured stromal cells (WPMY-1). Neurogenic, α1-adrenergic and non-adrenergic contractions were strongly inhibited with 50 µM and by around 50% with 10 µM genistein. Daidzein inhibited neurogenic contractions using 10 and 100 µM. Agonist-induced contractions were inhibited by 100 µM but not 10 µM daidzein. A combination of 6 µM genistein with 5 µM daidzein still inhibited neurogenic and agonist-induced contractions. Proliferation of WPMY-1 cells was inhibited by genistein (>50%) and daidzein (<50%). Genistein induced apoptosis and cell death (by seven-fold relative to controls), while daidzein induced cell death (6.4-fold) without apoptosis. Viability was reduced by genistein (maximum: 87%) and daidzein (62%). In conclusion, soy isoflavones exert sustained effects on prostate smooth muscle contractions and stromal cell growth, which may explain the inverse relationships between soy-rich nutrition, BPH and voiding symptoms.


Assuntos
Isoflavonas , Hiperplasia Prostática , Masculino , Humanos , Próstata/metabolismo , Genisteína/farmacologia , Adrenérgicos/metabolismo , Adrenérgicos/farmacologia , Músculo Liso , Contração Muscular , Hiperplasia Prostática/metabolismo , Células Estromais , Isoflavonas/farmacologia , Isoflavonas/metabolismo
5.
JCI Insight ; 7(22)2022 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-36219481

RESUMO

The fluid covering the surface of airway epithelia represents a first barrier against pathogens. The chemical and physical properties of the airway surface fluid are controlled by the activity of ion channels and transporters. In cystic fibrosis (CF), loss of CFTR chloride channel function causes airway surface dehydration, bacterial infection, and inflammation. We investigated the effects of IL-17A plus TNF-α, 2 cytokines with relevant roles in CF and other chronic lung diseases. Transcriptome analysis revealed a profound change with upregulation of several genes involved in ion transport, antibacterial defense, and neutrophil recruitment. At the functional level, bronchial epithelia treated in vitro with the cytokine combination showed upregulation of ENaC channel, ATP12A proton pump, ADRB2 ß-adrenergic receptor, and SLC26A4 anion exchanger. The overall result of IL-17A/TNF-α treatment was hyperviscosity of the airway surface, as demonstrated by fluorescence recovery after photobleaching (FRAP) experiments. Importantly, stimulation with a ß-adrenergic agonist switched airway surface to a low-viscosity state in non-CF but not in CF epithelia. Our study suggests that CF lung disease is sustained by a vicious cycle in which epithelia cannot exit from the hyperviscous state, thus perpetuating the proinflammatory airway surface condition.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística , Fibrose Cística , Humanos , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Depuração Mucociliar , Interleucina-17/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Adrenérgicos/farmacologia , Células Epiteliais/metabolismo , Fibrose Cística/genética , Citocinas/metabolismo , ATPase Trocadora de Hidrogênio-Potássio
6.
Nature ; 611(7934): 173-179, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36289326

RESUMO

G-protein-coupled receptors (GPCRs), the largest family of signalling receptors, as well as important drug targets, are known to activate extracellular-signal-regulated kinase (ERK)-a master regulator of cell proliferation and survival1. However, the precise mechanisms that underlie GPCR-mediated ERK activation are not clearly understood2-4. Here we investigated how spatially organized ß2-adrenergic receptor (ß2AR) signalling controls ERK. Using subcellularly targeted ERK activity biosensors5, we show that ß2AR signalling induces ERK activity at endosomes, but not at the plasma membrane. This pool of ERK activity depends on active, endosome-localized Gαs and requires ligand-stimulated ß2AR endocytosis. We further identify an endosomally localized non-canonical signalling axis comprising Gαs, RAF and mitogen-activated protein kinase kinase, resulting in endosomal ERK activity that propagates into the nucleus. Selective inhibition of endosomal ß2AR and Gαs signalling blunted nuclear ERK activity, MYC gene expression and cell proliferation. These results reveal a non-canonical mechanism for the spatial regulation of ERK through GPCR signalling and identify a functionally important endosomal signalling axis.


Assuntos
Adrenérgicos , Endossomos , MAP Quinases Reguladas por Sinal Extracelular , Receptores Adrenérgicos beta 2 , Adrenérgicos/metabolismo , Adrenérgicos/farmacologia , Proliferação de Células , Endossomos/efeitos dos fármacos , Endossomos/enzimologia , Endossomos/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Genes myc , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
7.
Life Sci ; 308: 120931, 2022 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-36084760

RESUMO

AIMS: Recently, the European Association of Urology recommended hexane-extracted fruit of Serenoa repens (HESr) in their guidelines on management of non-neurogenic male lower urinary tracts symptoms (LUTS). Despite previously lacking recommendations, Permixon® is the most investigated HESr in clinical trials, where it proved effective for male LUTS. In contrast, underlying mechanisms were rarely addressed and are only marginally understood. We therefore investigated effects of Permixon® on human prostate and detrusor smooth muscle contraction and on growth-related functions in prostate stromal cells. MAIN METHODS: Permixon® capsules were dissolved using n-hexane. Contractions of human prostate and detrusor tissues were induced in organ bath. Proliferation (EdU assay), growth (colony formation), apoptosis and cell death (flow cytometry), viability (CCK-8) and actin organization (phalloidin staining) were studied in cultured human prostate stromal cells (WPMY-1). KEY FINDINGS: Permixon® inhibited α1-adrenergic and thromboxane-induced contractions in prostate tissues, and methacholine-and thromboxane-induced contractions in detrusor tissues. Endothelin-1-induced contractions were not inhibited. Neurogenic contractions were inhibited in both tissues in a concentration-dependent manner. In WPMY-1 cells, Permixon® caused concentration-dependent breakdown of actin polymerization, inhibited colony formation, reduced cell viability, and proliferation, without showing cytotoxic or pro-apoptotic effects. SIGNIFICANCE: Our results provide a novel basis that allows, for the first time, to fully explain the ubiquitous beneficial effects of HESr in clinical trials. HESr may inhibit at least neurogenic, α1-adrenergic and thromboxane-induced smooth muscle contraction in the prostate and detrusor, and in parallel, prostate stromal cell growth. Together, this may explain symptom improvements by Permixon® in previous clinical trials.


Assuntos
Hiperplasia Prostática , Serenoa , Actinas/metabolismo , Adrenérgicos/farmacologia , Endotelina-1/metabolismo , Hexanos/metabolismo , Hexanos/farmacologia , Hexanos/uso terapêutico , Humanos , Masculino , Cloreto de Metacolina/metabolismo , Contração Muscular , Músculo Liso , Faloidina/metabolismo , Faloidina/farmacologia , Faloidina/uso terapêutico , Extratos Vegetais/uso terapêutico , Próstata/metabolismo , Hiperplasia Prostática/tratamento farmacológico , Hiperplasia Prostática/metabolismo , Sincalida/metabolismo , Células Estromais/metabolismo , Tromboxanos/metabolismo , Bexiga Urinária/metabolismo
8.
Toxicon ; 218: 57-65, 2022 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-36113683

RESUMO

Rhinella marina toad is abundant in Brazil. Its poison contains cardiac glycosides called bufadienolides, which are extensively investigated for their bioactivity. Our aim was to characterize the vasoactivity of Rhinella marina poison (RmP) on the aorta of male Wistar rats. For this, the RmP was first collected and processed to obtain an alcoholic extract. To determine cardiovascular effects of RmP, we performed in vivo tests by administering RmP intravenously in doses of 0.1-0.8 mg/kg. Vascular reactivity was also performed through concentration-response curves to RmP (10 ng/mL to 200 µg/mL) in aortic segments with and without endothelium. RmP induced a concentration-dependent contraction in rat aorta which was partly endothelium-mediated. Nitric oxide contributes with this response in view that incubation with L-NAME increased the contractile response. Additionally, treatment with indomethacin [cyclooxygenase, (COX) inhibitor], nifedipine (L-type voltage-gated calcium channels blocker), and BQ-123 (ETA receptors antagonist) decreased maximum response, and ketanserin (5-HT2 receptors antagonist) decreased pEC50, suggesting active participation of these pathways in the contractile response. On the other hand, apocynin (NADPH oxidase inhibitor) did not alter contractility. Incubation with prazosin (α1-adrenergic receptor antagonist) abolished the contractile response, suggesting that the RmP-induced contraction is dependent on the adrenergic pathway. In the Na+/K+ ATPase protocol, a higher Emax was observed in the RmP experimental group, suggesting that RmP potentiated Na+/K+ATPase hyperpolarizing response. When this extract was injected (i.v.) in vivo, increase in blood pressure and decrease in heart rate were observed. The results were immediate and transitory, and occurred in a dose-dependent manner. Overall, these data suggest that the poison extract of R. marina toad has an important vasoconstrictor action and subsequent vasopressor effects, and its use can be investigated to some cardiovascular disorders.


Assuntos
Bufanolídeos , Venenos , Adenosina Trifosfatases/metabolismo , Adenosina Trifosfatases/farmacologia , Adrenérgicos/farmacologia , Antagonistas Adrenérgicos/farmacologia , Animais , Bufanolídeos/toxicidade , Bufo marinus/metabolismo , Canais de Cálcio , Endotélio Vascular , Hemodinâmica , Indometacina/farmacologia , Ketanserina/farmacologia , Masculino , Metanol/farmacologia , NADPH Oxidases , NG-Nitroarginina Metil Éster , Nifedipino/farmacologia , Óxido Nítrico/metabolismo , Prazosina/farmacologia , Prostaglandina-Endoperóxido Sintases/metabolismo , Ratos , Ratos Wistar , Serotonina/farmacologia , Vasoconstritores
9.
Acta Physiol (Oxf) ; 236(3): e13881, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36039679

RESUMO

AIM: Cardiac autonomic nervous system (ANS) dysregulation is a hallmark of several cardiovascular diseases. Adrenergic signaling enhanced cardiomyocyte cohesion via PKA-mediated plakoglobin phosphorylation at serine 665, referred to as positive adhesiotropy. This study investigated cholinergic regulation of cardiomyocyte cohesion using muscarinic receptor agonist carbachol (CCH). METHODS: Dissociation assays, Western blot analysis, immunostaining, atomic force microscopy (AFM), immunoprecipitation, transmission electron microscopy (TEM), triton assays, and siRNA knockdown of genes were performed in either HL-1 cells or plakoglobin (PG) wild type (Jup+/+ ) and knockout (Jup-/- ) mice, which served as a model for arrhythmogenic cardiomyopathy. RESULTS: In HL-1 cells grown in norepinephrine (NE)-containing medium for baseline adrenergic stimulation, and murine cardiac slice cultures from Jup+/+ and Jup-/- mice CCH treatment impaired cardiomyocyte cohesion. Immunostainings and AFM experiments revealed that CCH reduced desmoglein 2 (DSG2) localization and binding at cell borders. Furthermore, CCH reduced intercalated disc plaque thickness in both Jup+/+ and Jup-/- mice, evidenced by TEM analysis. Immunoprecipitation experiments in HL-1 cells revealed no changes in DSG2 interaction with desmoplakin (DP), plakophilin 2 (PKP2), PG, and desmin (DES) after CCH treatment. However, knockdown of any of the above proteins abolished CCH-mediated loss of cardiomyocyte cohesion. Furthermore, in HL-1 cells, CCH inhibited adrenergic-stimulated ERK phosphorylation but not PG phosphorylation at serine 665. In addition, CCH activated the AKT/GSK-3ß axis in the presence of NE. CONCLUSION: Our results demonstrate that cholinergic signaling antagonizes the positive effect of adrenergic signaling on cardiomyocyte cohesion and thus causes negative adhesiotropy independent of PG phosphorylation.


Assuntos
Desmogleína 2 , Miócitos Cardíacos , Camundongos , Animais , Miócitos Cardíacos/metabolismo , Desmogleína 2/genética , Desmogleína 2/metabolismo , gama Catenina/metabolismo , gama Catenina/farmacologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Desmoplaquinas/metabolismo , Carbacol/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Placofilinas/metabolismo , RNA Interferente Pequeno/metabolismo , Desmina/metabolismo , Desmina/farmacologia , Colinérgicos/metabolismo , Colinérgicos/farmacologia , Receptores Muscarínicos/metabolismo , Adrenérgicos/farmacologia , Norepinefrina/metabolismo , Serina/metabolismo
10.
J Biochem Mol Toxicol ; 36(11): e23196, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35979984

RESUMO

Doxorubicin (DOX) is a potent chemotherapeutic agent used for cancer treatment, however, DOX-induced cardiotoxicity is a serious clinical problem because it causes acute and chronic heart dysfunction. Many studies have indicated that the α1-adrenergic receptor protects the heart from pathologic stress through activation survival signaling, however, the mechanism remains largely unknown. Previous studies have detected that the phenylephrine-induced complex-1 (PEX1) transcription factor, also known as zinc-finger protein 260 (Zfp260), is an effector of α1-adrenergic signaling in cardiac hypertrophy. Our present study aimed to investigate the role and underlying mechanism of PEX1 in cardiomyocyte survival during DOX-induced cardiotoxicity. Mice were exposed to a single intraperitoneal injection of DOX (15 mg/kg) to generate DOX-induced cardiotoxicity. We found that PEX1 expression was downregulated in DOX-treated murine hearts. PEX1 deficiency resulted in increased apoptosis, and conversely, PEX1 overexpression alleviated apoptosis induced by DOX in primary cardiomyocytes, as well as upregulated antiapoptotic genes such as BCL-2 and BCL-XL. Mechanistically, we identified that PEX1 might exert its antiapoptosis effect by playing a pivotal role in the action of α1-adrenergic signaling activation, which depends on the presence of GATA-4. Based on these findings, we supposed that PEX1 may be a novel transcription factor involved in cardiac cell survival and a promising candidate target for DOX-induced cardiotoxicity.


Assuntos
Adrenérgicos , Cardiotoxicidade , Camundongos , Animais , Cardiotoxicidade/metabolismo , Adrenérgicos/metabolismo , Adrenérgicos/farmacologia , Doxorrubicina/toxicidade , Miócitos Cardíacos/metabolismo , Apoptose , Fatores de Transcrição/metabolismo , Estresse Oxidativo , ATPases Associadas a Diversas Atividades Celulares/metabolismo , ATPases Associadas a Diversas Atividades Celulares/farmacologia
11.
Elife ; 112022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35913125

RESUMO

Ryanodine receptors (RyRs) exhibit dynamic arrangements in cardiomyocytes, and we previously showed that 'dispersion' of RyR clusters disrupts Ca2+ homeostasis during heart failure (HF) (Kolstad et al., eLife, 2018). Here, we investigated whether prolonged ß-adrenergic stimulation, a hallmark of HF, promotes RyR cluster dispersion and examined the underlying mechanisms. We observed that treatment of healthy rat cardiomyocytes with isoproterenol for 1 hr triggered progressive fragmentation of RyR clusters. Pharmacological inhibition of Ca2+/calmodulin-dependent protein kinase II (CaMKII) reversed these effects, while cluster dispersion was reproduced by specific activation of CaMKII, and in mice with constitutively active Ser2814-RyR. A similar role of protein kinase A (PKA) in promoting RyR cluster fragmentation was established by employing PKA activation or inhibition. Progressive cluster dispersion was linked to declining Ca2+ spark fidelity and magnitude, and slowed release kinetics from Ca2+ propagation between more numerous RyR clusters. In healthy cells, this served to dampen the stimulatory actions of ß-adrenergic stimulation over the longer term and protect against pro-arrhythmic Ca2+ waves. However, during HF, RyR dispersion was linked to impaired Ca2+ release. Thus, RyR localization and function are intimately linked via channel phosphorylation by both CaMKII and PKA, which, while finely tuned in healthy cardiomyocytes, underlies impaired cardiac function during pathology.


Assuntos
Insuficiência Cardíaca , Canal de Liberação de Cálcio do Receptor de Rianodina , Adrenérgicos/metabolismo , Adrenérgicos/farmacologia , Animais , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Insuficiência Cardíaca/metabolismo , Homeostase , Camundongos , Miócitos Cardíacos/metabolismo , Fosforilação , Ratos , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
12.
Clin Auton Res ; 32(4): 261-269, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35870087

RESUMO

PURPOSE: We investigate the impact of menopause on cardiovascular adjustments to the cold pressor test (CPT) and the role of the α1-adrenergic receptor. METHODS: Ten young women (YW) and nine postmenopausal women (MW) underwent 1 min of CPT in control and α1-blockade conditions (0.03 mg‧kg-1 of oral prazosin). RESULTS: CPT increased heart rate (HR) (YW: ∆20 ± 3 bpm; MW: ∆13 ± 2 bpm) and stroke volume (SV; YW: ∆15 ± 8 ml; MW: ∆9 ± 6 ml; p = 0.01 for time) and evoked a greater increase in cardiac output (CO) in YW (YW: ∆2.1 ± 0.2 l‧m-1; MW: ∆1.3 ± 0.5 l‧m-1; p = 0.01). α1-Blockade increased baseline HR and did not change HR, SV, and CO responses to CPT. MW presented an exaggerated systolic blood pressure (BP) response (YW: ∆38 ± 9 mmHg; MW: ∆56 ± 24 mmHg; p = 0.03). The α1-blockade did not change baseline BP while blunting its response. Total vascular resistance (TVR) was similar between groups at baseline and increased during CPT only in MW (YW: ∆2.3 ± 1.4 mmHg‧L-1‧min; MW:∆6.8 ± 5.9 mmHg‧L-1‧min). Under α1-blockade, the TVR increase during CPT was attenuated in MW and abolished in YW (YW: ∆0.3 ± 1.2 mmHg‧L-1‧min and MW: ∆3.0 ± 2.0 mmHg‧L-1‧min). CPT did not change femoral vascular conductance (FVC) in either group before the blockade (YW: ∆-0.3 ± 4.0 ml‧min-1‧mmHg-1; MW: ∆-0.2 ± 0.8 ml‧min-1‧mmHg-1); however, FVC tended to increase in young women (YW: ∆1.3 ± 1.0 ml‧min-1‧mmHg-1; MW: ∆0.1 ± 1.5 ml‧min-1‧mmHg-1; p = 0.06) after the α1-blockade. CONCLUSION: In postmenopausal women, the cardiac ability to adjust to CPT is blunted and α1-adrenergic receptor stimulation is important for the increase in stroke volume. In addition, the peripheral effect of α1-adrenergic receptor stimulation seems to be increased in postmenopausal women.


Assuntos
Sistema Cardiovascular , Sistema Nervoso Simpático , Adrenérgicos/farmacologia , Pressão Sanguínea/fisiologia , Temperatura Baixa , Feminino , Frequência Cardíaca/fisiologia , Humanos , Pós-Menopausa , Sistema Nervoso Simpático/fisiologia
13.
Basic Res Cardiol ; 117(1): 37, 2022 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-35842861

RESUMO

We have recently identified a pool of intracellular ß1 adrenergic receptors (ß1ARs) at the sarcoplasmic reticulum (SR) crucial for cardiac function. Here, we aim to characterize the integrative control of intracellular catecholamine for subcellular ß1AR signaling and cardiac function. Using anchored Förster resonance energy transfer (FRET) biosensors and transgenic mice, we determined the regulation of compartmentalized ß1AR-PKA signaling at the SR and plasma membrane (PM) microdomains by organic cation transporter 3 (OCT3) and monoamine oxidase A (MAO-A), two critical modulators of catecholamine uptake and homeostasis. Additionally, we examined local PKA substrate phosphorylation and excitation-contraction coupling in cardiomyocyte. Cardiac-specific deletion of MAO-A (MAO-A-CKO) elevates catecholamines and cAMP levels in the myocardium, baseline cardiac function, and adrenergic responses. Both MAO-A deletion and inhibitor (MAOi) selectively enhance the local ß1AR-PKA activity at the SR but not PM, and augment phosphorylation of phospholamban, Ca2+ cycling, and myocyte contractile response. Overexpression of MAO-A suppresses the SR-ß1AR-PKA activity and PKA phosphorylation. However, deletion or inhibition of OCT3 by corticosterone prevents the effects induced by MAOi and MAO-A deletion in cardiomyocytes. Deletion or inhibition of OCT3 also negates the effects of MAOi and MAO-A deficiency in cardiac function and adrenergic responses in vivo. Our data show that MAO-A and OCT3 act in concert to fine-tune the intracellular SR-ß1AR-PKA signaling and cardiac fight-or-flight response. We reveal a drug contraindication between anti-inflammatory corticosterone and anti-depressant MAOi in modulating adrenergic regulation in the heart, providing novel perspectives of these drugs with cardiac implications.


Assuntos
Corticosterona , Proteínas Quinases Dependentes de AMP Cíclico , Adrenérgicos/metabolismo , Adrenérgicos/farmacologia , Animais , Cálcio/metabolismo , Catecolaminas/metabolismo , Catecolaminas/farmacologia , Cátions/metabolismo , Cátions/farmacologia , Corticosterona/metabolismo , Corticosterona/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/farmacologia , Camundongos , Monoaminoxidase/metabolismo , Monoaminoxidase/farmacologia , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Fosforilação , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 1/metabolismo , Retículo Sarcoplasmático
14.
Mol Cell Endocrinol ; 549: 111619, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35337901

RESUMO

White adipocyte adiponectin exocytosis is triggered by cAMP and a concomitant increase of cytosolic Ca2+ potentiates its release. White adipose tissue is richly innervated by sympathetic nerves co-releasing noradrenaline (NA) and ATP, which may act on receptors in the adipocyte plasma membrane to increase cAMP via adrenergic receptors and Ca2+ via purinergic receptors. Here we determine the importance of NA and ATP for the regulation of white adipocyte adiponectin exocytosis, at the cellular and molecular level, and we specifically detail the ATP signalling pathway. We demonstrate that tyrosine hydroxylase (enzyme involved in catecholamine synthesis) is dramatically reduced in inguinal white adipose tissue (IWAT) isolated from mice with diet-induced obesity; this is associated with diminished levels of NA in IWAT and with a reduced ratio of high-molecular-weight (HMW) to total adiponectin in serum. Adiponectin exocytosis (measured as an increase in plasma membrane capacitance and as secreted product) is triggered by NA or ATP alone in cultured and primary mouse IWAT adipocytes, and enhanced by a combination of the two secretagogues. The ATP-induced adiponectin exocytosis is largely Ca2+-dependent and activated via purinergic P2Y2 receptors (P2Y2Rs) and the Gq11/PLC pathway. Adiponectin release induced by the nucleotide is abrogated in adipocytes isolated from obese and insulin-resistant mice, and this is associated with ∼70% reduced abundance of P2Y2Rs. The NA-triggered adiponectin exocytosis is likewise abolished in "obese adipocytes", concomitant with a 50% lower gene expression of beta 3 adrenergic receptors (ß3ARs). An increase in intracellular Ca2+ is not required for the NA-stimulated adiponectin secretion. Collectively, our data suggest that sympathetic innervation is a principal regulator of adiponectin exocytosis and that disruptions of this control are associated with the obesity-associated reduction of circulating levels of HMW/total adiponectin.


Assuntos
Adipócitos Brancos , Adiponectina , Trifosfato de Adenosina/metabolismo , Adipócitos Brancos/metabolismo , Adiponectina/metabolismo , Adrenérgicos/metabolismo , Adrenérgicos/farmacologia , Animais , Exocitose , Insulina/metabolismo , Camundongos , Norepinefrina/metabolismo , Norepinefrina/farmacologia , Obesidade/metabolismo
15.
Arch Physiol Biochem ; 128(2): 394-401, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31711314

RESUMO

Methylglyoxal (MG) is dicarbonyl aldehyde generated intracellularly from glucose and from some other compounds. Its increased formation is associated with several harmful consequences. In the present study, short-term effects of MG on metabolism of isolated rat adipocytes were determined. Insulin-induced lipogenesis was unchanged by MG. However, epinephrine-stimulated lipolysis was shown to be significantly reduced in adipocytes exposed to 200 µM MG. This inhibitory effect was similar in the presence of low and high concentrations of glucose, and also in the presence of alanine. However, MG failed to affect lipolysis induced by forskolin (activator of adenylate cyclase), dibutyryl-cAMP (activator of PKA) and DPCPX (adenosine A1 receptor antagonist). It was also revealed that lipolysis was unchanged by MG in fat cells pre-incubated with this compound, and then stimulated with epinephrine alone. Our results suggest that MG may impair ß-adrenergic signalling in rat adipocytes due to interaction with epinephrine, and thereby disturbs lipolysis.


Assuntos
Adrenérgicos , Aldeído Pirúvico , Adipócitos/metabolismo , Adrenérgicos/metabolismo , Adrenérgicos/farmacologia , Animais , Lipólise , Aldeído Pirúvico/farmacologia , Ratos , Ratos Wistar
16.
FEBS Lett ; 595(16): 2085-2098, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34197627

RESUMO

Brown and beige adipocytes dissipate energy by uncoupling protein 1 (UCP1)-dependent and UCP1-independent thermogenesis, which may be utilized to develop treatments against obesity. We have found that mRNA and protein expression of the alanine/serine/cysteine transporter-1 (ASC-1) was induced during adipocyte differentiation of human brown-prone deep neck and beige-competent subcutaneous neck progenitors, and SGBS preadipocytes. cAMP stimulation of differentiated adipocytes led to elevated uptake of serine, cysteine, and glycine, in parallel with increased oxygen consumption, augmented UCP1-dependent proton leak, increased creatine-driven substrate cycle-coupled respiration, and upregulation of thermogenesis marker genes and several respiratory complex subunits; these outcomes were impeded in the presence of the specific ASC-1 inhibitor, BMS-466442. Our data suggest that ASC-1-dependent consumption of serine, cysteine, and glycine is required for efficient thermogenic stimulation of human adipocytes.


Assuntos
Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Adrenérgicos/farmacologia , Sistema y+ de Transporte de Aminoácidos/metabolismo , Aminoácidos/metabolismo , Termogênese , Transporte Biológico/efeitos dos fármacos , Humanos , Termogênese/efeitos dos fármacos
17.
Int J Mol Sci ; 22(13)2021 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-34281286

RESUMO

Norepinephrine (NE) neurons and extracellular NE exert some protective effects against a variety of insults, including methamphetamine (Meth)-induced cell damage. The intimate mechanism of protection remains difficult to be analyzed in vivo. In fact, this may occur directly on target neurons or as the indirect consequence of NE-induced alterations in the activity of trans-synaptic loops. Therefore, to elude neuronal networks, which may contribute to these effects in vivo, the present study investigates whether NE still protects when directly applied to Meth-treated PC12 cells. Meth was selected based on its detrimental effects along various specific brain areas. The study shows that NE directly protects in vitro against Meth-induced cell damage. The present study indicates that such an effect fully depends on the activation of plasma membrane ß2-adrenergic receptors (ARs). Evidence indicates that ß2-ARs activation restores autophagy, which is impaired by Meth administration. This occurs via restoration of the autophagy flux and, as assessed by ultrastructural morphometry, by preventing the dissipation of microtubule-associated protein 1 light chain 3 (LC3) from autophagy vacuoles to the cytosol, which is produced instead during Meth toxicity. These findings may have an impact in a variety of degenerative conditions characterized by NE deficiency along with autophagy impairment.


Assuntos
Metanfetamina/antagonistas & inibidores , Metanfetamina/toxicidade , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Norepinefrina/farmacologia , Receptores Adrenérgicos beta 2/metabolismo , Adrenérgicos/farmacologia , Animais , Autofagia/efeitos dos fármacos , Compartimento Celular/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/administração & dosagem , Estimulantes do Sistema Nervoso Central/antagonistas & inibidores , Estimulantes do Sistema Nervoso Central/toxicidade , Desipramina/farmacologia , Relação Dose-Resposta a Droga , Metanfetamina/administração & dosagem , Microscopia Eletrônica de Transmissão , Modelos Neurológicos , Neurônios/ultraestrutura , Fármacos Neuroprotetores/farmacologia , Norepinefrina/metabolismo , Células PC12 , Ratos , Vacúolos/efeitos dos fármacos , Vacúolos/metabolismo , Vacúolos/ultraestrutura
18.
Metallomics ; 13(7)2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34185060

RESUMO

Copper (Cu) is an essential trace element that plays an important role in maintaining neuronal functions such as the biosynthesis of neurotransmitters. In contrast, exposure to excess Cu results in cell injury. Therefore, intracellular Cu levels are strictly regulated by proteins related to Cu-trafficking, including ATP7A. Parkinson's disease (PD) is a neurodegenerative disorder and is characterized by the loss of dopaminergic neurons in the substantia nigra. Recently, the abnormality of Cu homeostasis was demonstrated to be related to the pathogenesis of PD. However, the association between Cu dyshomeostasis and PD remains unclear. In this study, we examined the effects of 6-hydroxydopamine (6-OHDA), a neurotoxin used for the production of PD model animals, on cellular Cu trafficking in human neuroblastoma SH-SY5Y cells. 6-OHDA reduced the protein levels of the Cu exporter ATP7A and the Cu chaperone Atox1, but not CTR1, a Cu importer; however, it did not affect the expression of ATP7A and Atox1 mRNAs. The decreased levels of ATP7A and Atox1 proteins were restored by the antioxidant N-acetylcysteine and the lysosomal inhibitor bafilomycin A1. This suggests that 6-OHDA-induced oxidative stress facilitates the degradation of these proteins. In addition, the amount of intracellular Cu after exposure to CuCl2 was significantly higher in cells pretreated with 6-OHDA than in untreated cells. Moreover, 6-OHDA reduced the protein levels of the cuproenzyme dopamine ß-hydroxylase that converts dopamine to noradrenaline. Thus, this study suggests that 6-OHDA disrupts Cu homeostasis through the dysregulation of cellular Cu trafficking, resulting in the dysfunction of neuronal cells.


Assuntos
Proteínas de Transporte de Cobre/antagonistas & inibidores , ATPases Transportadoras de Cobre/antagonistas & inibidores , Cobre/metabolismo , Homeostase , Chaperonas Moleculares/antagonistas & inibidores , Neuroblastoma/patologia , Estresse Oxidativo , Oxidopamina/farmacologia , Adrenérgicos/farmacologia , Morte Celular , Humanos , Neuroblastoma/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Células Tumorais Cultivadas
19.
Meat Sci ; 177: 108490, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33744607

RESUMO

Muscle fiber morphometry and physicochemical characteristics were evaluated in LT muscles obtained from entire male lambs treated with zilpaterol hydrochloride (ZH, 0 and 0.15 mg/kg body weight) and/or steroidal implant (SI, with and without trenbolone acetate/estradiol). ZH and SI acted synergistically to increase LT area, type-IIb fiber cross-sectional area and soluble collagen content, likewise to decrease metmyoglobin concentration and insoluble collagen content. Ash content and ultimate pH showed a decrease due to an antagonistic effect between ZH and SI. Content of total collagen, protein, fat, moisture, oxidized lipids and water-holding capacity were unaffected by ZH and SI. Supplemental ZH, but not SI, decreased all color parameters and tended to increase shear force. Overall, the SI implantation of male lambs followed by a ZH supplementation promoted greater LT hypertrophy, without affecting protein and fat content, and physicochemical characteristics in their meat.


Assuntos
Fibras Musculares Esqueléticas/efeitos dos fármacos , Carne Vermelha/análise , Compostos de Trimetilsilil/farmacologia , Adrenérgicos/farmacologia , Animais , Cor , Estradiol/farmacologia , Masculino , Músculos Paraespinais/anatomia & histologia , Músculos Paraespinais/química , Resistência ao Cisalhamento , Carneiro Doméstico , Acetato de Trembolona/farmacologia
20.
Nat Commun ; 11(1): 1821, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32286326

RESUMO

The abscopal effect following ionizing radiation therapy (RT) is considered to be a rare event. This effect does occur more frequently when combined with other therapies, including immunotherapy. Here we demonstrate that the frequency of abscopal events following RT alone is highly dependent upon the degree of adrenergic stress in the tumor-bearing host. Using a combination of physiologic, pharmacologic and genetic strategies, we observe improvements in the control of both irradiated and non-irradiated distant tumors, including metastatic tumors, when adrenergic stress or signaling through ß-adrenergic receptor is reduced. Further, we observe cellular and molecular evidence of improved, antigen-specific, anti-tumor immune responses which also depend upon T cell egress from draining lymph nodes. These data suggest that blockade of ß2 adrenergic stress signaling could be a useful, safe, and feasible strategy to improve efficacy in cancer patients undergoing radiation therapy.


Assuntos
Adrenérgicos/farmacologia , Imunidade , Neoplasias/imunologia , Neoplasias/radioterapia , Radiação Ionizante , Estresse Fisiológico , Antagonistas Adrenérgicos beta/farmacologia , Animais , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Linfonodos/patologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias/genética , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA