Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 153
Filtrar
1.
Cardiovasc Res ; 117(1): 137-148, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31995173

RESUMO

AIMS: Cardiac sympathetic overactivation is an important trigger of ventricular arrhythmias in patients with chronic heart failure (CHF). Our previous study demonstrated that N-type calcium (Cav2.2) currents in cardiac sympathetic post-ganglionic (CSP) neurons were increased in CHF. This study investigated the contribution of Cav2.2 channels in cardiac sympathetic overactivation and ventricular arrhythmogenesis in CHF. METHODS AND RESULTS: Rat CHF was induced by surgical ligation of the left coronary artery. Lentiviral Cav2.2-α shRNA or scrambled shRNA was transfected in vivo into stellate ganglia (SG) in CHF rats. Final experiments were performed at 14 weeks after coronary artery ligation. Real-time polymerase chain reaction and western blot data showed that in vivo transfection of Cav2.2-α shRNA reduced the expression of Cav2.2-α mRNA and protein in the SG in CHF rats. Cav2.2-α shRNA also reduced Cav2.2 currents and cell excitability of CSP neurons and attenuated cardiac sympathetic nerve activities (CSNA) in CHF rats. The power spectral analysis of heart rate variability (HRV) further revealed that transfection of Cav2.2-α shRNA in the SG normalized CHF-caused cardiac sympathetic overactivation in conscious rats. Twenty-four-hour continuous telemetry electrocardiogram recording revealed that this Cav2.2-α shRNA not only decreased incidence and duration of ventricular tachycardia/ventricular fibrillation but also improved CHF-induced heterogeneity of ventricular electrical activity in conscious CHF rats. Cav2.2-α shRNA also decreased susceptibility to ventricular arrhythmias in anaesthetized CHF rats. However, Cav2.2-α shRNA failed to improve CHF-induced cardiac contractile dysfunction. Scrambled shRNA did not affect Cav2.2 currents and cell excitability of CSP neurons, CSNA, HRV, and ventricular arrhythmogenesis in CHF rats. CONCLUSIONS: Overactivation of Cav2.2 channels in CSP neurons contributes to cardiac sympathetic hyperactivation and ventricular arrhythmogenesis in CHF. This suggests that discovering purely selective and potent small-molecule Cav2.2 channel blockers could be a potential therapeutic strategy to decrease fatal ventricular arrhythmias in CHF.


Assuntos
Canais de Cálcio Tipo N/metabolismo , Insuficiência Cardíaca/metabolismo , Coração/inervação , Interferência de RNA , Gânglio Estrelado/metabolismo , Fibras Simpáticas Pós-Ganglionares/metabolismo , Taquicardia Ventricular/prevenção & controle , Fibrilação Ventricular/prevenção & controle , Potenciais de Ação , Animais , Cálcio/metabolismo , Canais de Cálcio Tipo N/genética , Sinalização do Cálcio , Células Cultivadas , Modelos Animais de Doenças , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/fisiopatologia , Frequência Cardíaca , Masculino , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos Sprague-Dawley , Gânglio Estrelado/fisiopatologia , Fibras Simpáticas Pós-Ganglionares/fisiopatologia , Taquicardia Ventricular/genética , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/fisiopatologia , Fibrilação Ventricular/genética , Fibrilação Ventricular/metabolismo , Fibrilação Ventricular/fisiopatologia
2.
Physiol Res ; 68(2): 209-217, 2019 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-30628826

RESUMO

To determine the influence of IGF-1 deletion on renal sympathetic nerve activity (RSNA), left ventricular dysfunction, and renal function in deoxycorticosterone acetate (DOCA)-salt hypertensive mice. The DOCA-salt hypertensive mice models were constructed and the experiment was classified into WT (Wild-type mice) +sham, LID (Liver-specific IGF-1 deficient mice) + sham, WT + DOCA, and LID+ DOCA groups. Enzyme-linked immunosorbent assay (ELISA) was used to detect the serum IGF-1 levels in mice. The plasma norepinephrine (NE), urine protein, urea nitrogen and creatinine, as well as RSNA were measured. Echocardiography was performed to assess left ventricular dysfunction, and HE staining to observe the pathological changes in renal tissue of mice. DOCA-salt induction time-dependently increased the systolic blood pressure (SBP) of mice, especially in DOCA-salt LID mice. Besides, the serum IGF-1 levels in WT mice were decreased after DOCA-salt induction. In addition, the plasma NE concentration and NE spillover, urinary protein, urea nitrogen, creatinine and RSNA were remarkably elevated with severe left ventricular dysfunction, but the creatinine clearance was reduced in DOCA-salt mice, and these similar changes were obvious in DOCA-salt mice with IGF-1 deletion. Moreover, the DOCA-salt mice had tubular ectasia, glomerular fibrosis, interstitial cell infiltration, and increased arterial wall thickness, and the DOCA-salt LID mice were more serious in those aspects. Deletion of IGF-1 may lead to enhanced RSNA in DOCA-salt hypertensive mice, thereby further aggravating left ventricular dysfunction and renal damage.


Assuntos
Acetato de Desoxicorticosterona/toxicidade , Hipertensão/sangue , Fator de Crescimento Insulin-Like I/deficiência , Rim/fisiologia , Fibras Simpáticas Pós-Ganglionares/metabolismo , Disfunção Ventricular Esquerda/sangue , Animais , Hipertensão/induzido quimicamente , Hipertensão/fisiopatologia , Fator de Crescimento Insulin-Like I/genética , Rim/efeitos dos fármacos , Rim/inervação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mineralocorticoides/toxicidade , Norepinefrina/sangue , Fibras Simpáticas Pós-Ganglionares/efeitos dos fármacos , Disfunção Ventricular Esquerda/induzido quimicamente , Disfunção Ventricular Esquerda/fisiopatologia
3.
Physiol Rep ; 6(18): e13877, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30230240

RESUMO

The astrocytic glutamate transporter (GLT1) plays an important role in the maintenance of extracellular glutamate concentration below neurotoxic levels in brain. However, the functional role of GLT1 within the nucleus of the solitary tract (NTS) in the regulation of cardiovascular function remains unclear. We examined the effect of inhibiting GLT1 in the subpostremal NTS on mean arterial pressure (MAP), renal sympathetic nerve activity (RSNA) and heart rate (HR) in anesthetized, artificially ventilated rats. It was found that dihydrokainate (DHK; inhibitor of GLT1, 5 mmol/L, 100 nL) injections into the NTS (n = 6) decreased MAP (50 ± 10 mmHg, mean ± SD), RSNA (89 ± 14%) and HR (37 ± 6 bpm). Pretreatment with kynurenate (KYN; glutamate receptor antagonist, 5 mmol/L, 30 µL) topically applied to the dorsal surface of the brainstem (n = 4) attenuated the responses to NTS injections of DHK (P < 0.01). The effect of DHK on arterial baroreflex function was examined using i.v. infusions of phenylephrine and nitroprusside. DHK reduced baroreflex response range (maximum-minimum) of RSNA by 91 ± 2% and HR by 83 ± 5% (n = 6, P < 0.001). These results indicate that inhibition of GLT1 within the NTS decreases MAP, RSNA, and HR by the activation of ionotropic glutamate receptors. As a result, baroreflex control of RSNA and HR was dramatically attenuated. The astrocytic glutamate transporter in the NTS plays an important role in the maintenance and regulation of cardiovascular function.


Assuntos
Barorreflexo/fisiologia , Transportador 2 de Aminoácido Excitatório/antagonistas & inibidores , Frequência Cardíaca/fisiologia , Núcleo Solitário/metabolismo , Fibras Simpáticas Pós-Ganglionares/metabolismo , Animais , Barorreflexo/efeitos dos fármacos , Transportador 2 de Aminoácido Excitatório/metabolismo , Frequência Cardíaca/efeitos dos fármacos , Ácido Caínico/administração & dosagem , Ácido Caínico/análogos & derivados , Masculino , Microinjeções/métodos , Ratos , Ratos Sprague-Dawley , Núcleo Solitário/efeitos dos fármacos , Fibras Simpáticas Pós-Ganglionares/efeitos dos fármacos , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/metabolismo
4.
Neurosci Lett ; 684: 18-24, 2018 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-29966754

RESUMO

The autonomic nervous system innervates various peripheral tissue functions. Various external stimuli affect autonomic nerve activity, however, there is little information about the involvement of sensory receptors in the responses. The TRPA1 is a calcium-permeable non-selective cation channel which plays a crucial role in the susceptibility to various stimuli. ß-Eudesmol, an oxygenated sesquiterpene found in hop essential oil and beer, activates the TRPA1. Intragastric administration of ß-eudesmol decreased efferent adrenal sympathetic nerve activity (ASNA) in rats, whereas subcutaneous administration did not. ASNA suppression by ß-eudesmol was not observed in TRPA1 knockout rats. The ß-eudesmol derived ASNA suppression was partially, but significantly, eliminated by subdiaphragmatic vagotomy in rats, suggesting the afferent vagal nerve from the gastrointestinal tract to the brain is involved in the effect of ß-eudesmol on ASNA. Our results indicate that ß-eudesmol suppresses ASNA, partly through TRPA1 and the afferent vagus nerve. These findings introduce the physiological significance of the TRPA1 in the control of ASNA.


Assuntos
Glândulas Suprarrenais/inervação , Glândulas Suprarrenais/metabolismo , Sesquiterpenos de Eudesmano/farmacologia , Fibras Simpáticas Pós-Ganglionares/metabolismo , Sistema Nervoso Simpático/metabolismo , Canal de Cátion TRPA1/deficiência , Glândulas Suprarrenais/efeitos dos fármacos , Animais , Vias Eferentes/efeitos dos fármacos , Vias Eferentes/metabolismo , Epinefrina/metabolismo , Masculino , Ratos , Ratos Transgênicos , Ratos Wistar , Sesquiterpenos de Eudesmano/química , Fibras Simpáticas Pós-Ganglionares/efeitos dos fármacos , Sistema Nervoso Simpático/efeitos dos fármacos
5.
Int Heart J ; 58(5): 787-793, 2017 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-28966311

RESUMO

A large amount of norepinephrine (NE) released from cardiac sympathetic nerve terminals might accelerate myocardial ischemic injury. Nicorandil (NICO), KATP channel opener, could attenuate cardiac NE release from the sympathetic nerve terminals during ischemia. The present study aimed to investigate the effects of NICO-induced attenuation of cardiac NE release on myocardial ischemia-reperfusion (I/R) injury in rats, by comparison with the effect of cardiac sympathetic denervation on I/R injury.Cardiac interstitial NE (iNE) concentrations were determined using a microdialysis method. Rats were divided into 3 groups; control, NICO, and denervation groups. Cardiac sympathetic denervation was performed by painting 10% phenol on the left ventricular epicardium 7 days before producing ischemia. The left coronary artery was ligated for 30 minutes and then re-perfused for 120 minutes. NICO (50 µg/kg/minute) was infused intravenously starting 20 minutes before the coronary occlusion to the end of the ligation.The infarct size of the left ventricle was smaller in rats treated with NICO than in control rats (20.2 ± 3.0 versus 50.6 ± 14.7%, P < 0.01). Sympathetic denervation also reduced infarct size (28.5 ± 10.4 %, P < 0.01), which was not significantly different from that in the NICO group. At the end of 30-minute ischemia, iNE increased markedly in control rats (0.1 ± 0.1 to 20.6 ± 5.3 × 103 pg/mL), whereas the increase was completely inhibited in denervated rats. NICO markedly attenuated the increase (4.9 ± 3.0 × 103 pg/mL, P < 0.01) during ischemia.NICO-induced attenuation of neural NE release during ischemia might, at least in part, contribute to myocardial protection against I/R injury.


Assuntos
Ventrículos do Coração/inervação , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Miocárdio/metabolismo , Nicorandil/farmacologia , Norepinefrina/antagonistas & inibidores , Fibras Simpáticas Pós-Ganglionares/metabolismo , Animais , Modelos Animais de Doenças , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Masculino , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Norepinefrina/metabolismo , Ratos , Ratos Wistar , Fibras Simpáticas Pós-Ganglionares/efeitos dos fármacos , Complexo Vitamínico B/farmacologia
6.
Brain Res ; 1602: 111-8, 2015 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-25625357

RESUMO

Pelvic ganglion (PG) neurons relay sympathetic and parasympathetic signals to the lower urinary tract, comprising the urinary bladder and bladder outlet, and are thus essential for both storage and voiding reflexes. Autonomic transmission is mediated by activation of the nicotinic acetylcholine receptor (nAChR) in PG neurons. Previously, bladder outlet obstruction (BOO), secondary to benign prostatic hyperplasia, was found to increase soma sizes of bladder-projecting PG neurons. To date, however, it remains unknown whether these morphological changes are accompanied by functional plasticity in PG neurons. In the present study, we investigated whether BOO alters acetylcholine receptor (nAChR) transcript expression and current density in bladder PG neurons. Partial ligation of the rat urethra for six weeks induced detrusor overactivity (DO), as observed during cystometrical measurement. In rats exhibiting DO, membrane capacitance of parasympathetic bladder PG neurons was selectively increased. Real-time PCR analysis revealed that BOO enhanced the expression of the transcripts encoding the nAChR α3 and ß4 subunits in PG neurons. Notably, BOO significantly increased ACh-evoked current density in parasympathetic bladder PG neurons, whereas no changes were observed in sympathetic bladder and parasympathetic penile PG neurons. In addition, other ligand-gated ionic currents were immune to BOO in bladder PG neurons. Taken together, these data suggest that BOO causes upregulation of nAChR in parasympathetic bladder PG neurons, which in turn may potentiate ganglionic transmission and contribute to the development of DO.


Assuntos
Neurônios/metabolismo , Receptores Nicotínicos/metabolismo , Obstrução do Colo da Bexiga Urinária/metabolismo , Bexiga Urinária Hiperativa/metabolismo , Bexiga Urinária/diagnóstico por imagem , Animais , Membrana Celular/fisiologia , Modelos Animais de Doenças , Capacitância Elétrica , Masculino , Técnicas de Rastreamento Neuroanatômico , Neurônios/patologia , Fibras Parassimpáticas Pós-Ganglionares/metabolismo , Fibras Parassimpáticas Pós-Ganglionares/patologia , Técnicas de Patch-Clamp , Pênis/inervação , Cintilografia , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Fibras Simpáticas Pós-Ganglionares/metabolismo , Fibras Simpáticas Pós-Ganglionares/patologia , Regulação para Cima , Bexiga Urinária/patologia , Obstrução do Colo da Bexiga Urinária/patologia , Bexiga Urinária Hiperativa/patologia
7.
J Neurochem ; 131(2): 219-28, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24848581

RESUMO

In Parkinson's disease (PD), profound putamen dopamine (DA) depletion reflects denervation and a shift from vesicular sequestration to oxidative deamination of cytoplasmic DA in residual terminals. PD also involves cardiac sympathetic denervation. Whether PD entails myocardial norepinephrine (NE) depletion and a sequestration-deamination shift have been unknown. We measured apical myocardial tissue concentrations of NE, DA, and their neuronal metabolites 3,4-dihydroxyphenylglycol (DHPG), and 3,4-dihydroxyphenylacetic acid (DOPAC) from 23 PD patients and 23 controls and ascertained the extent of myocardial NE depletion in PD. We devised, validated in VMAT2-Lo mice, and applied 5 neurochemical indices of the sequestration-deamination shift-concentration ratios of DOPAC:DA, DA:NE, DHPG:NE, DOPAC:NE, and DHPG:DOPAC-and used a kinetic model to estimate the extent of the vesicular storage defect. The PD group had decreased myocardial NE content (p < 0.0001). The majority of patients (70%) had severe NE depletion (mean 2% of control), and in this subgroup all five indices of a sequestration-deamination shift were increased compared to controls (p < 0.001 for each). Vesicular storage in residual nerves was estimated to be decreased by 84-91% in this subgroup. We conclude that most PD patients have severe myocardial NE depletion, because of both sympathetic denervation and decreased vesicular storage in residual nerves. We found that the majority (70%) of Parkinson's disease (PD) patients have profound (98%) myocardial norepinephrine depletion, because of both cardiac sympathetic denervation and a shift from vesicular sequestration to oxidative deamination of cytoplasmic catecholamines in the residual nerves. This shift may be part of a final common pathogenetic pathway in the loss of catecholaminergic neurons that characterizes PD.


Assuntos
Miocárdio/metabolismo , Estresse Oxidativo/fisiologia , Doença de Parkinson/metabolismo , Fibras Simpáticas Pós-Ganglionares/metabolismo , Vesículas Sinápticas/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Desaminação/fisiologia , Dopamina/metabolismo , Feminino , Humanos , Masculino , Camundongos , Miocárdio/patologia , Norepinefrina/metabolismo , Doença de Parkinson/patologia , Fibras Simpáticas Pós-Ganglionares/patologia , Proteínas Vesiculares de Transporte de Monoamina/deficiência , Proteínas Vesiculares de Transporte de Monoamina/metabolismo
8.
Acta Histochem ; 116(1): 48-54, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23791475

RESUMO

Developmental expression of Wilms' tumor gene (WT1) and protein is crucial for cell proliferation, apoptosis, differentiation and cytoskeletal architecture regulation. Recently, a potential role of WT1 has been suggested in the development of neural tissue and in neurodegenerative disorders. We have investigated immunohistochemically the developmentally regulated expression and distribution of WT1 in the human fetal peripheral sympathetic nervous system (PSNS) and the gastro-enteric nervous system (GENS) from weeks 8 to 28 gestational age. WT1 expression was restricted to the cytoplasm of sympathetic neuroblasts, while it progressively disappeared with advancing morphologic differentiation of these cells along both ganglionic and chromaffin cell lineages. In adult tissues, both ganglion and chromaffin cells lacked any WT1 expression. These findings show that WT1 is a reliable marker of human sympathetic neuroblasts, which can be used routinely in formalin-fixed, paraffin-embedded tissues. The progressive loss of WT1 in both ganglion and chromaffin cells, suggests its potential repressor role of differentiation in a precise temporal window during the development of the human PSNS and GENS.


Assuntos
Trato Gastrointestinal/inervação , Fibras Simpáticas Pós-Ganglionares/metabolismo , Proteínas WT1/metabolismo , Células Cromafins/metabolismo , Trato Gastrointestinal/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Imuno-Histoquímica , Células-Tronco Neurais/metabolismo , Especificidade de Órgãos , Fibras Simpáticas Pós-Ganglionares/embriologia , Proteínas WT1/genética
9.
Cardiovasc Toxicol ; 13(4): 381-90, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23877628

RESUMO

Angiotensin II type 1 receptor (AT1-R) and nuclear factor-kappaB (NF-κB) in the paraventricular nucleus (PVN) play important roles in heart failure (HF); however, the central mechanisms by which AT1-R and NF-κB contribute to sympathoexcitation in HF are yet unclear. In this study, we determined whether interaction between AT1-R and NF-κB in the PVN modulates neurotransmitters and contributes to NAD(P)H oxidase-dependent oxidative stress and sympathoexcitation in HF. Rats were implanted with bilateral PVN cannulae and subjected to coronary artery ligation or sham surgery (SHAM). Subsequently, animals were treated for 4 weeks through bilateral PVN infusion with either vehicle or losartan (LOS, 10 µg/h), an AT1-R antagonist; or pyrrolidine dithiocarbamate (PDTC, 5 µg/h), a NF-κB inhibitor via osmotic minipump. Myocardial infarction (MI) rats had higher levels of glutamate (Glu), norepinephrine (NE) and NF-κB p65 activity, lower levels of gamma-aminobutyric acid (GABA), and more positive neurons for phosphorylated IKKß and gp91(phox) (a subunit of NAD(P)H oxidase) in the PVN when compared to SHAM rats. MI rats also had higher levels of renal sympathetic nerve activity (RSNA) and plasma proinflammatory cytokines (PICs), NE and epinephrine. PVN infusions of LOS or PDTC attenuated the decreases in GABA and the increases in gp91(phox), NF-κB activity, Glu and NE, in the PVN of HF rats. PVN infusions of LOS or PDTC also attenuated the increases in RSNA and plasma PICs, NE and epinephrine in MI rats. These findings suggest that interaction between AT1 receptor and NF-κB in the PVN contributes to oxidative stress and sympathoexcitation by modulating neurotransmitters in heart failure.


Assuntos
Insuficiência Cardíaca/metabolismo , NF-kappa B/metabolismo , Estresse Oxidativo/fisiologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Fibras Simpáticas Pós-Ganglionares/metabolismo , Animais , Losartan/farmacologia , Masculino , NF-kappa B/antagonistas & inibidores , Neurotransmissores/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Ligação Proteica/fisiologia , Pirrolidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Fibras Simpáticas Pós-Ganglionares/efeitos dos fármacos , Tiocarbamatos/farmacologia
10.
J Neurosci ; 33(17): 7175-83, 2013 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-23616527

RESUMO

Sympathetic nerves can regenerate after injury to reinnervate target tissues. Sympathetic regeneration is well documented after chronic cardiac ischemia, so we were surprised that the cardiac infarct remained denervated following ischemia-reperfusion (I-R). We used mice to ask if the lack of sympathetic regeneration into the scar was due to blockade by inhibitory extracellular matrix within the infarct. We found that chondroitin sulfate proteoglycans (CSPGs) were present in the infarct after I-R, but not after chronic ischemia, and that CSPGs caused inhibition of sympathetic axon outgrowth in vitro. Ventricle explants after I-R and chronic ischemia stimulated sympathetic axon outgrowth that was blocked by nerve growth factor antibodies. However, growth in I-R cocultures was asymmetrical, with axons growing toward the heart tissue consistently shorter than axons growing in other directions. Growth toward I-R explants was rescued by adding chondroitinase ABC to the cocultures, suggesting that I-R infarct-derived CSPGs prevented axon extension. Sympathetic ganglia lacking protein tyrosine phosphatase sigma (PTPRS) were not inhibited by CSPGs or I-R explants in vitro, suggesting PTPRS is the major CSPG receptor in sympathetic neurons. To test directly if infarct-derived CSPGs prevented cardiac reinnervation, we performed I-R in ptprs-/- and ptprs+/- mice. Cardiac infarcts in ptprs-/- mice were hyperinnervated, while infarcts in ptprs+/- littermates were denervated, confirming that CSPGs prevent sympathetic reinnervation of the cardiac scar after I-R. This is the first example of CSPGs preventing sympathetic reinnervation of an autonomic target following injury, and may have important consequences for cardiac function and arrhythmia susceptibility after myocardial infarction.


Assuntos
Proteoglicanas de Sulfatos de Condroitina/fisiologia , Coração/inervação , Coração/fisiopatologia , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Fibras Simpáticas Pós-Ganglionares/metabolismo , Animais , Técnicas de Cocultura , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Infarto do Miocárdio/fisiopatologia , Técnicas de Cultura de Órgãos , Fibras Simpáticas Pós-Ganglionares/fisiopatologia
11.
Auton Neurosci ; 177(2): 95-100, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23506793

RESUMO

Gastric leptin elicits its cardiovascular and splanchnic sympathoinhibitory responses via a vagal afferent mechanism, however the latter are blunted/abolished in animals fed a medium high fat diet (MHFD). In a diet-induced obesity model we sought to determine whether the renal sympathetic nerve discharge (RSND) and regional vasodilator responses to gastric leptin are also affected by diet and/or obesity. The diet induced obesity model was used in 2 separate studies. After 13 weeks on a MHFD the animals were classified as either obesity prone (OP) or obesity resistant (OR) depending on their weight gain. Control animals were fed a low fat diet for an equivalent period. Arterial pressure (AP) and heart rate (HR) were monitored in isoflurane-anaesthetised, artificially ventilated animals and RSND or regional vascular responses to leptin (15 µg/kg) administered close to the coeliac artery were evaluated. OP rats had higher baseline AP compared to control/OR rats (P<0.05). Close arterial leptin inhibited RSND in control animals but this response was abolished in OR and OP animals (P<0.01 for both). Leptin administration increased renal vascular conductance in control animals but this response was significantly attenuated only in OP animals (P<0.05). The vasodilator response in the superior mesenteric artery was not significantly different in any of the groups (P>0.05). Together these results suggest that, while the renal sympathoinhibitory responses to gastric leptin are affected by diet, the vasodilator responses to leptin in the renal vascular bed are only affected in OP animals. These changes may impact on cardiovascular homeostatic mechanisms in obesity.


Assuntos
Dieta Hiperlipídica , Rim/irrigação sanguínea , Rim/inervação , Leptina/sangue , Obesidade/sangue , Fibras Simpáticas Pós-Ganglionares/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Artéria Celíaca/efeitos dos fármacos , Artéria Celíaca/metabolismo , Dieta com Restrição de Gorduras/métodos , Dieta Hiperlipídica/métodos , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Rim/efeitos dos fármacos , Leptina/administração & dosagem , Masculino , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Obesidade/prevenção & controle , Ratos , Ratos Sprague-Dawley , Fibras Simpáticas Pós-Ganglionares/efeitos dos fármacos , Resultado do Tratamento , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia
12.
PLoS One ; 7(10): e48119, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23118937

RESUMO

Our group recently demonstrated that maternal high-fat diet (HFD) consumption is associated with non-alcoholic fatty liver disease, increased apoptosis, and changes in gluconeogenic gene expression and chromatin structure in fetal nonhuman primate (NHP) liver. However, little is known about the long-term effects that a HFD has on hepatic nervous system development in offspring, a system that plays an important role in regulating hepatic metabolism. Utilizing immunohistochemistry and Real-Time PCR, we quantified sympathetic nerve fiber density, apoptosis, inflammation, and other autonomic components in the livers of fetal and one-year old Japanese macaques chronically exposed to a HFD. We found that HFD exposure in-utero and throughout the postnatal period (HFD/HFD), when compared to animals receiving a CTR diet for the same developmental period (CTR/CTR), is associated with a 1.7 fold decrease in periportal sympathetic innervation, a 5 fold decrease in parenchymal sympathetic innervation, and a 2.5 fold increase in hepatic apoptosis in the livers of one-year old male animals. Additionally, we observed an increase in hepatic inflammation and a decrease in a key component of the cholinergic anti-inflammatory pathway in one-year old HFD/HFD offspring. Taken together, these findings reinforce the impact that continuous exposure to a HFD has in the development of long-term hepatic pathologies in offspring and highlights a potential neuroanatomical basis for hepatic metabolic dysfunction.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Fígado/inervação , Efeitos Tardios da Exposição Pré-Natal/etiologia , Sistema Nervoso Simpático/embriologia , Animais , Apoptose , Citocinas/genética , Citocinas/metabolismo , Feminino , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Transportador de Glucose Tipo 2/genética , Transportador de Glucose Tipo 2/metabolismo , Glicogênio/metabolismo , Hepatite/etiologia , Hepatite/metabolismo , Hepatite/patologia , Mediadores da Inflamação/metabolismo , Fígado/embriologia , Fígado/crescimento & desenvolvimento , Fígado/metabolismo , Macaca , Masculino , Troca Materno-Fetal , Neuropeptídeo Y/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/patologia , Receptores de Neuropeptídeo Y/genética , Receptores de Neuropeptídeo Y/metabolismo , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Fibras Simpáticas Pós-Ganglionares/metabolismo , Fibras Simpáticas Pós-Ganglionares/patologia , Sistema Nervoso Simpático/crescimento & desenvolvimento , Sistema Nervoso Simpático/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
13.
J Pharmacol Exp Ther ; 343(1): 97-105, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22761303

RESUMO

During myocardial ischemia/reperfusion, lipid peroxidation leads to the formation of toxic aldehydes that contribute to ischemic dysfunction. Mitochondrial aldehyde dehydrogenase type 2 (ALDH2) alleviates ischemic heart damage and reperfusion arrhythmias via aldehyde detoxification. Because excessive norepinephrine release in the heart is a pivotal arrhythmogenic mechanism, we hypothesized that neuronal ALDH2 activation might diminish ischemic norepinephrine release. Incubation of cardiac sympathetic nerve endings with acetaldehyde, at concentrations achieved in myocardial ischemia, caused a concentration-dependent increase in norepinephrine release. A major increase in norepinephrine release also occurred when sympathetic nerve endings were incubated in hypoxic conditions. ALDH2 activation substantially reduced acetaldehyde- and hypoxia-induced norepinephrine release, an action prevented by inhibition of ALDH2 or protein kinase Cε (PKCε). Selective activation of G(i/o)-coupled adenosine A(1), A(3), or histamine H(3) receptors markedly inhibited both acetaldehyde- and hypoxia-induced norepinephrine release. These effects were also abolished by PKCε and/or ALDH2 inhibition. Moreover, A(1)-, A(3)-, or H(3)-receptor activation increased ALDH2 activity in a sympathetic neuron model (differentiated PC12 cells stably transfected with H(3) receptors). This action was prevented by the inhibition of PKCε and ALDH2. Our findings suggest the existence in sympathetic neurons of a protective pathway initiated by A(1)-, A(3)-, and H(3)-receptor activation by adenosine and histamine released in close proximity of these terminals. This pathway comprises the sequential activation of PKCε and ALDH2, culminating in aldehyde detoxification and inhibition of hypoxic norepinephrine release. Thus, pharmacological activation of PKCε and ALDH2 in cardiac sympathetic nerves may have significant protective effects by alleviating norepinephrine-induced life-threatening arrhythmias that characterize myocardial ischemia/reperfusion.


Assuntos
Aldeído Desidrogenase/metabolismo , Proteínas Mitocondriais/metabolismo , Isquemia Miocárdica/metabolismo , Norepinefrina/metabolismo , Proteína Quinase C-épsilon/fisiologia , Receptores Histamínicos/metabolismo , Receptores Purinérgicos P1/metabolismo , Aldeído-Desidrogenase Mitocondrial , Animais , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Cobaias , Hipóxia/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Norepinefrina/antagonistas & inibidores , Células PC12 , Ratos , Fibras Simpáticas Pós-Ganglionares/efeitos dos fármacos , Fibras Simpáticas Pós-Ganglionares/metabolismo , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo
14.
Curr Hypertens Rep ; 14(1): 38-45, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22068338

RESUMO

Catecholamines (epinephrine and norepinephrine) are synthesised and produced by the adrenal medulla and postganglionic nerve fibres of the sympathetic nervous system. It is known that essential hypertension has a significant neurogenic component, with the rise in blood pressure mediated at least in part by overactivity of the sympathetic nervous system. Moreover, novel therapeutic strategies aimed at reducing sympathetic activity show promise in the treatment of hypertension. This article reviews recent advances within this rapidly changing field, particularly focusing on the role of genetic polymorphisms within key catecholamine biosynthetic enzymes, cofactors, and storage molecules. In addition, mechanisms linking the sympathetic nervous system and other adverse cardiovascular states (obesity, insulin resistance, dyslipidaemia) are discussed, along with speculation as to how recent scientific advances may lead to the emergence of novel antihypertensive treatments.


Assuntos
Anti-Hipertensivos , Pressão Sanguínea , Catecolaminas , Hipertensão , Fibras Simpáticas Pós-Ganglionares , Medula Suprarrenal/metabolismo , Medula Suprarrenal/fisiopatologia , Anti-Hipertensivos/metabolismo , Anti-Hipertensivos/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/genética , Catecolaminas/biossíntese , Catecolaminas/genética , Catecolaminas/metabolismo , Cromograninas/genética , Cromograninas/metabolismo , Coenzimas/genética , Coenzimas/metabolismo , Descoberta de Drogas , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Hipertensão/tratamento farmacológico , Hipertensão/genética , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Polimorfismo Genético , Fibras Simpáticas Pós-Ganglionares/efeitos dos fármacos , Fibras Simpáticas Pós-Ganglionares/metabolismo , Fibras Simpáticas Pós-Ganglionares/fisiopatologia , Terapias em Estudo , Canais de Potencial de Receptor Transitório/genética , Canais de Potencial de Receptor Transitório/metabolismo
15.
Ann Pathol ; 31(5): 396-8, 2011 Oct.
Artigo em Francês | MEDLINE | ID: mdl-21982249

RESUMO

Oncocytic metaplasia of the nasopharynx is an exceptional lesion which exact etiopathogenesis, although largely discussed, still remains controversial. The purpose of this paper is to present the epidemiological characteristics and clinical signs of this lesion and to study its pathogenesis and its therapeutic modalities. We report two cases that occurred respectively in a 53- and 60-year-old woman. The first presented with pharyngeal dysesthesia and otalgia. The endoscopic examination revealed an irregularity of the posterior wall of the nasopharynx. The second patient presented with tinnitus, discomfort of the left ear and bilateral hearing loss. Endoscopic exam revealed a bilateral structural abnormality to the eardrum. Microscopy showed focal oncocytic metaplasia of the nasopharynx mucosa in both cases. There was a positive outcare for both patients after excisional biopsy. Oncocytic metaplasia seems to be in relation to the stimulation of sympathic neuropeptidergic nerve fibers which target epithelial, connective, endothelial and lymphoid cells.


Assuntos
Nasofaringe/patologia , Células Oxífilas/patologia , Biópsia , Transtornos de Deglutição/etiologia , Dor de Orelha/etiologia , Células Epiteliais/patologia , Feminino , Perda Auditiva Bilateral/etiologia , Humanos , Metaplasia , Pessoa de Meia-Idade , Mucosa Nasal/patologia , Nasofaringe/cirurgia , Neuropeptídeos/metabolismo , Pseudolinfoma/etiologia , Fibras Simpáticas Pós-Ganglionares/metabolismo , Zumbido/etiologia
16.
Auton Neurosci ; 164(1-2): 43-50, 2011 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-21724473

RESUMO

Current evidence indicates that rises in systemic levels of estrogen create in the uterus an inhibitory environment for sympathetic nerves. However, molecular insights of these changes are far from complete. We evaluated if semaphorin 3F mRNA, a sympathetic nerve repellent, was produced by the rat uterus and if its expression was modulated by estrogen. We also analyzed whether uterine nerves express the semaphorin 3F binding receptor, neuropilin-2. Uterine levels of semaphorin 3F mRNA were measured using real time reverse transcriptase-polymerase chain reaction in prepubertal rat controls and following chronic estrogen treatment. Localization of semaphorin 3F transcripts was determined by in situ hybridization and the expression of neuropilin-2 was assessed by immunohistochemistry. These studies showed that: (1) chronic estrogen treatment led to a 5-fold induction of semaphorin 3F mRNA in the immature uterus; (2) estrogen provoked a tissue-specific induction of semaphorin 3F which was particularly localized in the connective tissue that borders muscle bundles and surrounds intrauterine blood vessels; (3) two major cell-types were recognized in the areas where transcripts were concentrated, fibroblast-like cells and infiltrating eosinophil leukocytes; and (4) some delicate nerve terminal profiles present in the estrogenized uterus were immunoreactive for neuropilin-2. Temporal and spatial expression patterns of semaphorin 3F/neuropilin-2 are consistent with a possible role of this guidance cue in the remodeling of uterine sympathetic innervation by estrogen. Though correlative in its nature, these data support a model whereby semaphorin 3F, in combination with other inhibitory molecules, converts the estrogenized myometrium to an inhospitable environment for sympathetic nerves.


Assuntos
Estrogênios/fisiologia , Miométrio/inervação , Degeneração Neural/metabolismo , Degeneração Neural/fisiopatologia , Proteínas do Tecido Nervoso/biossíntese , Fibras Simpáticas Pós-Ganglionares/metabolismo , Regulação para Cima/fisiologia , Útero/inervação , Animais , Feminino , Peptídeos e Proteínas de Sinalização Intracelular/agonistas , Peptídeos e Proteínas de Sinalização Intracelular/genética , Miométrio/fisiologia , Proteínas do Tecido Nervoso/agonistas , Proteínas do Tecido Nervoso/genética , Ratos , Ratos Wistar , Útero/fisiologia
17.
Auton Neurosci ; 164(1-2): 13-9, 2011 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-21646052

RESUMO

Cardiac sympathetic neurons stimulate heart rate and the force of contraction through release of norepinephrine. Nerve growth factor modulates sympathetic transmission through activation of TrkA and p75NTR. Nerve growth factor plays an important role in post-infarct sympathetic remodeling. We used mice lacking p75NTR to examine the effect of altered nerve growth factor signaling on sympathetic neuropeptide expression, cardiac norepinephrine, and ventricular function after myocardial infarction. Infarct size was similar in wildtype and p75NTR-/- mice after ischemia-reperfusion surgery. Likewise, mRNAs encoding vasoactive intestinal peptide, galanin, and pituitary adenylate cyclase activating peptides were identical in wildtype and p75NTR-/- cardiac sympathetic neurons, as was expression of the TrkA neurotrophin receptor. Norepinephrine content was elevated in the base of the p75NTR-/- ventricle compared to wildtype, but levels were identical below the site of occlusion. Left ventricular pressure, dP/dt(MAX), and dP/dt(MIN) were measured under isoflurane anesthesia 3 and 7 days after surgery. Ventricular pressure decreased significantly 3 days after infarction, and deficits in dP/dt(MAX) were revealed by stimulating beta receptors with dobutamine and release of endogenous norepinephrine with tyramine. dP/dt(MIN) was not altered by genotype or surgical group. Few differences were observed between genotypes 3 days after surgery, in contrast to low pressure and dP/dt(MAX) previously reported in control p75NTR-/- animals. Seven days after surgery ventricular pressure and dP/dt(MAX) were significantly lower in p75NTR-/- hearts compared to WT hearts. Thus, the lack of p75NTR did not enhance cardiac function after myocardial infarction.


Assuntos
Ventrículos do Coração/inervação , Ventrículos do Coração/fisiopatologia , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/fisiopatologia , Norepinefrina/metabolismo , Receptores de Fator de Crescimento Neural/fisiologia , Fibras Simpáticas Pós-Ganglionares/fisiopatologia , Animais , Modelos Animais de Doenças , Feminino , Ventrículos do Coração/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/genética , Norepinefrina/biossíntese , Norepinefrina/fisiologia , Receptores de Fator de Crescimento Neural/deficiência , Receptores de Fator de Crescimento Neural/genética , Fibras Simpáticas Pós-Ganglionares/metabolismo , Regulação para Cima/genética
18.
Exp Mol Pathol ; 91(1): 353-61, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21545801

RESUMO

Elevated levels of endogenous estrogens occurring in the course of pathological states of ovaries (follicular cysts, tumors) as well as xenoestrogens may result in hyperestrogenism. In rat, a close relationship between estrogens and sympathetic and sensory neurons supplying the genito-urinary system was reported. Recently, we have shown that long-term estradiol-17ß (E(2)) administration affected morphological and immunochemical organization of the sympathetic ovarian neurons in the caudal mesenteric ganglion of adult gilts. In this study, the influence of E(2) overdose on the number and distribution of neurons in the sympathetic chain ganglia (SChG) projecting to the ovary of adult pigs was investigated. The numbers of ovarian dopamine-ß-hydroxylase (DßH-), neuropeptide Y (NPY-), somatostatin (SOM-), galanin (GAL-) and estrogen receptors (ERs-) immunoreactive perikarya as well as the density of the intraganglionic nerve fibers containing DßH and/or NPY, SOM, GAL were also determined. On day 3 of the estrous cycle the ovaries of both the control and experimental gilts were injected with retrograde neuronal tracer Fast Blue, to identify the neurons innervating gonads. From day 4 of the estrous cycle to the expected day 20 of the second studied cycle, the experimental gilts were injected with E(2), while the control gilts were receiving oil. After the last E(2)/oil injection, the SChG Th16-S2 were collected and processed for double-labeling immunofluorescence. Injections of E(2): (1) increased the E(2) level in the peripheral blood ~4-5 fold, (2) reduced the total number of Fast Blue-positive postganglionic neurons in the ganglia under investigation, (3) decreased the number of perikarya in the L2-L4 ganglia, (4) reduced the number of perikarya in the ventral, dorsal and central regions of the SChG, (5) decreased the numbers of DßH(+)/NPY(+) and DßH(+)/GAL(+) perikarya and the numbers of DßH(+) but NPY(-), SOM(-) and GAL(-) perikarya in the SChG, (6) decreased the number of perikarya expressing ERs subtype α and ß, and (7) decreased the total number of the intraganglionic nerve fibers containing DßH and/or NPY. These results show that long-term E(2) treatment of adult gilts down-regulates the population of both noradrenergic and ERs expressing the SChG ovary supplying neurons. Our findings suggest also that elevated E(2) levels that occur during pathological states may regulate gonadal function(s) by affecting ovary supplying neurons.


Assuntos
Estradiol/farmacologia , Gânglios Simpáticos/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Ovário/inervação , Animais , Biomarcadores/metabolismo , Estradiol/sangue , Feminino , Gânglios Simpáticos/patologia , Neurônios/metabolismo , Neurônios/patologia , Ovário/efeitos dos fármacos , Suínos , Fibras Simpáticas Pós-Ganglionares/efeitos dos fármacos , Fibras Simpáticas Pós-Ganglionares/metabolismo , Fibras Simpáticas Pós-Ganglionares/patologia
20.
Acta Physiol (Oxf) ; 203(1): 271-8, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21276205

RESUMO

AIM: Vascular production of hydrogen peroxide (H(2)O(2)) is implicated in the development and progression of vascular disease. Hydrogen peroxide also promotes neuronal degeneration, which suggests that vascular H(2)O(2) would promote degeneration of perivascular sympathetic nerves. Vascular cells also produce vascular endothelial growth factor (VEGF), which could protect perivascular nerves from the detrimental effects of H(2)O(2) . The aim of this study was to test these hypotheses. METHODS: The effects of H(2)O(2) and VEGF on neuronal survival and noradrenaline uptake were studied in cultures of rat post-ganglionic sympathetic neurones. Western analyses of catalase and growth associated protein 43 were performed and reactive oxygen species (ROS) were measured using the fluorescent indicator 5-(and-6)-chloromethyl-2'7'-dichlorodihydrofluorescein diacetate, acetyl ester. RESULTS: Hydrogen peroxide (30 µm) decreased the survival of post-ganglionic sympathetic neurones (57.8 ± 4.8% of control) and decreased noradrenaline uptake into the neurones (14 ± 6% of control). Hyperglycaemia, which is known to increase H(2)O(2), also decreased survival (31.4 ± 12% of control) and noradrenaline uptake (42 ± 18.4% of control). VEGF reduced the effects of H(2)O(2) (94.3 ± 12% of control) and hyperglycaemia (83.5 ± 23.6% of control) on survival. VEGF increased catalase, a primary determinant of intracellular concentrations of H(2)O(2) , and decreased H(2)O(2) -induced increases in ROS. CONCLUSION: These results indicate that VEGF protects post-ganglionic sympathetic neurones from the detrimental effects of H(2)O(2). Our data suggest that an increase in catalase is the mechanisms underlying this neuroprotection.


Assuntos
Catalase/biossíntese , Peróxido de Hidrogênio/toxicidade , Oxidantes/toxicidade , Fibras Simpáticas Pós-Ganglionares/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Fibras Simpáticas Pós-Ganglionares/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...