Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Braz. J. Psychiatry (São Paulo, 1999, Impr.) ; 44(1): 6-14, Jan.-Feb. 2022. tab, graf
Artigo em Inglês | LILACS-Express | LILACS | ID: biblio-1360181

RESUMO

Objective: Electroconvulsive therapy (ECT) is a well-established therapeutic intervention for major depressive disorder. Recent literature has shown that the anesthetic agent ketamine has some antidepressant properties at low doses and may be an alternative therapy for treatment-resistant major depressive disorder. We hypothesized that the use of low-dose ketamine as an anesthetic adjunct in ECT would more rapidly improve depression while maintaining hemodynamic stability than ECT with propofol alone. Methods: Institutional ethics approval was obtained, and the use of ketamine in this study was approved by Health Canada. This is a randomized, double-blinded, placebo-controlled trial that involved ketamine administration at 0.5 mg/kg IV in addition to propofol anesthesia for ECT. The primary outcome was the number of ECT treatments required to achieve a 50% reduction in the Montgomery-Asberg Depression Rating Scale (MADRS). Secondary outcomes included the number of ECT treatments required to achieve a 25% reduction in MADRS score, as well as any differences in the Clinical Global Impression Scale for Severity, hemodynamic variables, and seizure duration. Adverse events were recorded for safety assessment. Results: A total of 45 patients completed the study. No difference was found between groups with respect to the primary or secondary outcomes. The ketamine group showed a trend towards a decreased dose of propofol required to achieve adequate anesthesia. No adverse events were reported. Conclusion: Low-dose ketamine does not improve psychiatric outcomes in the setting of propofol-based anesthesia for ECT. Specifically, ketamine did not reduce the number of ECT sessions necessary to achieve a 50 or 25% reduction in MADRS scores. Reassuringly, the fact that no differences in hemodynamic variables or unexpected adverse events occurred suggests that low-dose ketamine may be safely used in this setting should clinical indications warrant its use. Clinical trial registration: ClinicalTrials.gov, NCT02579642

2.
Cardiovasc Res ; 116(1): 183-192, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30715197

RESUMO

AIMS: Perinatal iron deficiency (ID) alters developmental trajectories of offspring, predisposing them to cardiovascular dysfunction in later life. The mechanisms underlying this long-term programming of renal function have not been defined. We hypothesized perinatal ID causes hypertension and alters kidney metabolic function and morphology in a sex-dependent manner in adult offspring. Furthermore, we hypothesized these effects are exacerbated by chronic consumption of a high salt diet. METHODS AND RESULTS: Pregnant Sprague Dawley rats were fed either an iron-restricted or replete diet prior to and throughout pregnancy. Adult offspring were fed normal or high salt diets for 6 weeks prior to experimentation at 6 months of age. Blood pressure (BP) was assessed via indwelling catheters in anaesthetized offspring; kidney mitochondrial function was assessed via high-resolution respirometry; reactive oxygen species and nitric oxide were quantified via fluorescence microscopy. Adult males, but not females, exhibited increased systolic BP due to ID (P = 0.01) and high salt intake (P = 0.02). In males, but not in females, medullary mitochondrial content was increased by high salt (P = 0.003), while succinate-dependent respiration was reduced by ID (P < 0.05). The combination of perinatal ID and high salt reduced complex IV activity in the cortex of males (P = 0.01). Perinatal ID increased cytosolic superoxide generation (P < 0.001) concomitant with reduced nitric oxide bioavailability (P < 0.001) in male offspring, while high salt increased mitochondrial superoxide in the medulla (P = 0.04) and cytosolic superoxide within the cortex (P = 0.01). Male offspring exhibited glomerular basement membrane thickening (P < 0.05), increased collagen deposition (P < 0.05), and glomerular hypertrophy (interaction, P = 0.02) due to both perinatal ID and high salt. Female offspring exhibited no alterations in mitochondrial function or morphology due to either high salt or ID. CONCLUSION: Perinatal ID causes long-term sex-dependent alterations in renal metabolic function and morphology, potentially contributing to hypertension and increased cardiovascular disease risk.


Assuntos
Deficiências de Ferro , Ferro da Dieta , Nefropatias/etiologia , Rim/metabolismo , Mitocôndrias/metabolismo , Estresse Oxidativo , Efeitos Tardios da Exposição Pré-Natal , Sódio na Dieta , Fatores Etários , Fenômenos Fisiológicos da Nutrição Animal , Animais , Pressão Sanguínea , Modelos Animais de Doenças , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Feminino , Rim/patologia , Rim/fisiopatologia , Nefropatias/metabolismo , Nefropatias/patologia , Nefropatias/fisiopatologia , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Mitocôndrias/patologia , Óxido Nítrico/metabolismo , Estado Nutricional , Gravidez , Ratos Sprague-Dawley , Fatores Sexuais , Superóxidos/metabolismo , Fatores de Tempo
3.
J Physiol ; 597(18): 4715-4728, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31368136

RESUMO

KEY POINTS: Perinatal iron deficiency causes changes in offspring mesenteric artery function in adulthood, particularly in males, which can be exacerbated by chronic intake of a high salt diet. Perinatal iron deficient male offspring exhibit enhanced conversion of big endothelin-1 to active endothelin-1, coinciding with decreased nitric oxide levels. Perinatal iron deficient male offspring have reduced nitric oxide-mediated endothelial-dependent vasodilatation coincident with increased vascular superoxide levels following consumption of a high salt diet. Perinatal iron deficiency has no apparent effects on vascular function in female offspring, even when fed a high salt diet. These results help us better understand underlying vascular mechanisms contributing to increased cardiovascular risk from perinatal stressors such as iron deficiency. ABSTRACT: Pre- and immediate postnatal stressors, such as iron deficiency, can alter developmental trajectories and predispose offspring to long-term cardiovascular dysfunction. Here, we investigated the impact of perinatal iron deficiency on vascular function in the adult offspring, and whether these long-term effects were exacerbated by prolonged consumption of a high salt diet in adulthood. Female Sprague Dawley rats were fed either an iron-restricted or -replete diet prior to and throughout pregnancy. Six weeks prior to experimentation at 6 months of age, adult offspring were fed either a normal or high salt diet. Mesenteric artery responses to vasodilators and vasoconstrictors were assessed ex vivo by wire myography. Male perinatal iron deficient offspring exhibited decreased reliance on nitric oxide with methacholine-induced vasodilatation (interaction P = 0.03), coincident with increased superoxide levels when fed the high salt diet (P = 0.01). Male perinatal iron deficient offspring exhibit enhanced big endothelin-1 conversion to active endothelin-1 (P = 0.02) concomitant with decreased nitric oxide levels (P = 0.005). Female offspring vascular function was unaffected by perinatal iron deficiency, albeit the high salt diet was associated with impaired vasodilation and decreased nitric oxide production (P = 0.02), particularly in the perinatal iron deficient offspring. These findings implicate vascular dysfunction in the sex-specific programming of cardiovascular dysfunction in the offspring by perinatal iron deficiency.


Assuntos
Anemia Ferropriva/fisiopatologia , Dieta/efeitos adversos , Endotélio Vascular/efeitos dos fármacos , Parto/efeitos dos fármacos , Cloreto de Sódio na Dieta/farmacologia , Doenças Vasculares/induzido quimicamente , Animais , Endotélio Vascular/metabolismo , Feminino , Masculino , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/metabolismo , Óxido Nítrico/metabolismo , Gravidez , Ratos , Ratos Sprague-Dawley , Doenças Vasculares/metabolismo , Vasoconstritores/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
4.
FASEB J ; 32(6): 3254-3263, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29401611

RESUMO

Prenatal iron deficiency alters fetal developmental trajectories, which results in persistent changes in organ function. Here, we studied the effects of prenatal iron deficiency on fetal kidney and liver mitochondrial function. Pregnant Sprague-Dawley rats were fed partially or fully iron-restricted diets to induce a state of moderate or severe iron deficiency alongside iron-replete control rats. We assessed mitochondrial function via high-resolution respirometry and reactive oxygen species generation via fluorescence microscopy on gestational d 21. Hemoglobin levels were reduced in dams in the moderate (-31%) and severe groups (-54%) compared with controls, which was accompanied by 55% reductions in fetal hemoglobin levels in both moderate and severe groups versus controls. Male iron-deficient kidneys exhibited globally reduced mitochondrial content and respiration, as well as increased cytosolic superoxide and decreased NO. Female iron-deficient kidneys exhibited complex II down-regulation and increased mitochondrial oxidative stress. Male iron-deficient livers exhibited reduced complex IV respiration and increased cytosolic superoxide, whereas female liver tissues exhibited no alteration in oxidant levels or mitochondrial function. These findings indicate that prenatal iron deficiency causes changes in mitochondrial content and function as well as oxidant status in a sex- and organ-dependent manner, which may be an important mechanism that underlies the programming of cardiovascular disease.-Woodman, A. G., Mah, R., Keddie, D., Noble, R. M. N., Panahi, S., Gragasin, F. S., Lemieux, H., Bourque, S. L. Prenatal iron deficiency causes sex-dependent mitochondrial dysfunction and oxidative stress in fetal rat kidneys and liver.


Assuntos
Feto/metabolismo , Deficiências de Ferro , Rim/embriologia , Fígado/embriologia , Mitocôndrias Hepáticas/metabolismo , Estresse Oxidativo , Complicações na Gravidez/metabolismo , Caracteres Sexuais , Animais , Feminino , Feto/patologia , Rim/patologia , Fígado/patologia , Masculino , Mitocôndrias Hepáticas/patologia , Gravidez , Complicações na Gravidez/patologia , Ratos , Ratos Sprague-Dawley
5.
Sci Rep ; 7: 46573, 2017 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-28440316

RESUMO

Prenatal iron-deficiency (ID) is known to alter fetal developmental trajectories, which predisposes the offspring to chronic disease in later life, although the underlying mechanisms remain unclear. Here, we sought to determine whether varying degrees of maternal anaemia could induce organ-specific patterns of hypoxia in the fetuses. Pregnant female Sprague Dawley rats were fed iron-restricted or iron-replete diets to induce a state of moderate (M-ID) or severe ID (S-ID) alongside respective controls. Ultrasound biomicroscopy was performed on gestational day (GD)20 to assess uterine and umbilical artery blood flow patterns. On GD21, tissues were collected and assessed for hypoxia using pimonidazole staining. Compared to controls, maternal haemoglobin (Hb) in M- and S-ID were reduced 17% (P < 0.01) and 48% (P < 0.001), corresponding to 39% (P < 0.001) and 65% (P < 0.001) decreases in fetal Hb. Prenatal ID caused asymmetric fetal growth restriction, which was most pronounced in S-ID. In both severities of ID, umbilical artery resistive index was increased (P < 0.01), while pulsatility index only increased in S-ID (P < 0.05). In both M-and S-ID, fetal kidneys and livers showed evidence of hypoxia (P < 0.01 vs. controls), whereas fetal brains and placentae remained normoxic. These findings indicate prenatal ID causes organ-specific fetal hypoxia, even in the absence of severe maternal anaemia.


Assuntos
Anemia Ferropriva , Encéfalo , Doenças Fetais/sangue , Deficiências de Ferro , Placenta , Anemia Ferropriva/sangue , Anemia Ferropriva/embriologia , Anemia Ferropriva/patologia , Animais , Encéfalo/irrigação sanguínea , Encéfalo/embriologia , Encéfalo/patologia , Feminino , Placenta/irrigação sanguínea , Placenta/embriologia , Placenta/patologia , Gravidez , Ratos , Ratos Sprague-Dawley
6.
Can J Anaesth ; 64(4): 361-369, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28070833

RESUMO

PURPOSE: To determine if a non-exercise algorithm-derived assessment of cardiorespiratory fitness (CRFA) accurately predicted estimated values obtained using a six-minute walk test (CRF6MWD) and the Duke Activity Status Index (CRFDASI). METHODS: Following research ethics board approval, an observational cohort study was conducted in selected, consenting patients undergoing elective surgery. Participants completed questionnaires assessing their self-reported exercise capacity. Their height, weight, waist circumference, and vital signs were measured. A six-minute walk test was performed twice with a 45-min rest interval between tests. The correlation between CRFA and both CRF6MWD and CRFDASI was determined. RESULTS: Two hundred forty-two participants were included. Mean age was 62 (range 45-88 yr); 150 (62%) were male, 87 (36%) self-reported walking or jogging > 16 km per week, and 49 (20%) were current smokers. The CRFA and CRF6MWD were highly correlated (Pearson r = 0.878; P < 0.001). CRFA and CRFDASI were less strongly correlated (Pearson r = 0.252; P < 0.001). Among patients capable of walking > 427 m in the six-minute walk test, CRFA, CRF6MWD, and CRFDASI were equivalent. CONCLUSION: A non-exercise algorithm can estimate cardiorespiratory fitness in patients presenting for elective surgery. The variables required to compute CRFA can be obtained in a clinic setting without the need to engage in formal exercise testing. Further evaluation of CRFA as a predictor of long-term outcome in patients is warranted.


Assuntos
Aptidão Cardiorrespiratória/fisiologia , Teste de Esforço/métodos , Cuidados Pré-Operatórios/métodos , Inquéritos e Questionários , Idoso , Idoso de 80 Anos ou mais , Algoritmos , Estudos de Coortes , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Reprodutibilidade dos Testes , Procedimentos Cirúrgicos Operatórios
7.
A A Case Rep ; 6(6): 160-2, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26825990

RESUMO

Pseudotumor cerebri syndrome (PTCS) is a rare disorder chiefly observed in obese women of childbearing age. We describe a case of a parturient with PTCS managed successfully with an intrathecal catheter, after inadvertent dural puncture, for labor analgesia, surgical anesthesia, and treatment of headache because of intracranial hypertension during the peripartum period. Prolonged placement of the intrathecal catheter (i.e., >24 hours) may have contributed to the absence of postdural puncture headache symptoms and an uneventful postpartum period. Intrathecal catheter placement may therefore be a viable option in patients with PTCS should inadvertent dural puncture occur.


Assuntos
Analgesia Obstétrica/métodos , Cateterismo/instrumentação , Pseudotumor Cerebral/tratamento farmacológico , Adulto , Parto Obstétrico , Feminino , Humanos , Injeções Espinhais , Cefaleia Pós-Punção Dural/prevenção & controle , Gravidez
8.
Drugs Aging ; 22(10): 823-44, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16245957

RESUMO

The historical basis for understanding erectile function as a neurovascular phenomenon and the advance from fanciful to effective treatment of erectile dysfunction (ED) are reviewed, with emphasis on patients with cardiovascular disease (CVD). ED occurs in 60% of CVD patients by 40 years of age. Male ED and female sexual dysfunction (FSD) diminish quality of life and often warn of occult CVD. ED is often unrecognised but is readily diagnosed during a 5-minute interview using a truncated International Index of Erectile Function questionnaire. Erection of the penis and clitoral engorgement result from local, arousal-induced release of neuronal and endothelial-derived nitric oxide (NO). Arterial vasodilatation and relaxation of cavernosal smooth muscle cells cause arterial blood to flood trabecular spaces, compressing venous drainage, resulting in tumescence. Cyclic guanosine monophosphate (cGMP)-induced activation of protein kinase G mediates the effects of NO by enhancing calcium sequestration and activating large-conductance, calcium-sensitive K+ channels. Future treatment strategies will likely enhance these pathways. Phosphodiesterase-5 inhibitors (sildenafil, tadalafil and vardenafil) increase cGMP levels in erectile tissue. These agents are effective in 80% of CVD patients with ED and can be used safely, even in the presence of stable coronary disease or congestive heart failure, provided nitrates are avoided and patients do not have hypotension, severe aortic stenosis or evocable myocardial ischaemia. Second-line therapies (vacuum constrictor device and transurethral or intracavernosal prostaglandin E1) can also be used in CVD patients. Treatment of FSD and its relationship to CVD are less well established, but similarities to ED exist. ED can be prevented by reduction of CVD risk factors, exercise, weight loss and abstinence from smoking.


Assuntos
Doenças Cardiovasculares/complicações , Inibidores de Fosfodiesterase , Diester Fosfórico Hidrolases/metabolismo , Disfunções Sexuais Psicogênicas , 3',5'-GMP Cíclico Fosfodiesterases , Doenças Cardiovasculares/enzimologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5 , Disfunção Erétil/enzimologia , Disfunção Erétil/etiologia , Disfunção Erétil/terapia , Feminino , Humanos , Masculino , Inibidores de Fosfodiesterase/administração & dosagem , Inibidores de Fosfodiesterase/efeitos adversos , Inibidores de Fosfodiesterase/uso terapêutico , Fatores de Risco , Disfunções Sexuais Psicogênicas/enzimologia , Disfunções Sexuais Psicogênicas/etiologia , Disfunções Sexuais Psicogênicas/terapia
9.
FASEB J ; 18(12): 1382-91, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15333581

RESUMO

Female sexual function is under-studied, and mechanisms of clitoral engorgement-relaxation are incompletely understood. Penile erection results from nitric oxide (NO) -induced cyclic guanosine monophosphate (cGMP) accumulation. cGMP-dependent protein kinase (PKG) activates large-conductance, calcium-activated potassium channels (BK(Ca)), thereby hyperpolarizing and relaxing vascular and trabecular smooth muscle cells, allowing engorgement. We hypothesize rat clitorises relax by a similar mechanism. Rat clitorises express components of the proposed pathway: neuronal and endothelial NO synthases, soluble guanylyl cyclase (sGC), type 5 phosphodiesterase (PDE-5), and BK(Ca) channels. The NO donor diethylamine NONOate (DEANO), the PKG activator 8-pCPT-cGMP, and the PDE-5 inhibitor sildenafil, cause dose-dependent clitoral relaxation that is inhibited by antagonists of PKG (Rp-8-Br-cGMPS) or BK(Ca) channels (iberiotoxin). Electrical field stimulation induces tetrodotoxin-sensitive NO release and relaxation that is inhibited by the Na+ channel blocker tetrodotoxin or sGC inhibitor 1H-(1,2,4)oxadiozolo(4,3-a)quinoxalin-1-one. Human BK(Ca) channels, transferred to Chinese hamster ovary cells via an adenoviral vector, and endogenous rat clitoral smooth muscle K+ current are activated by this PKG-dependent mechanism. Laser confocal microscopy reveals protein expression of BK(Ca) channels on clitoral smooth muscle cells; these cells exhibit BK(Ca) channel activity that is activated by both DEANO and sildenafil. We conclude that neurovascular derived NO causes clitoral relaxation via a PKG-dependent activation of BK(Ca) channels. The BK(Ca) channel is an appealing target for drug therapy of female erectile dysfunction.


Assuntos
Clitóris/fisiologia , GMP Cíclico/metabolismo , Óxido Nítrico/metabolismo , Canais de Potássio Cálcio-Ativados/metabolismo , Animais , Células CHO , Cálcio/metabolismo , Clitóris/anatomia & histologia , Clitóris/irrigação sanguínea , Clitóris/inervação , Cricetinae , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Estimulação Elétrica , Eletrofisiologia , Feminino , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Canais de Potássio Ativados por Cálcio de Condutância Alta , Lasers , Microdissecção , Relaxamento Muscular/efeitos dos fármacos , Músculo Liso/irrigação sanguínea , Músculo Liso/efeitos dos fármacos , Músculo Liso/inervação , Músculo Liso/fisiologia , Óxido Nítrico/biossíntese , Piperazinas/farmacologia , Canais de Potássio Cálcio-Ativados/genética , Purinas , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Citrato de Sildenafila , Sulfonas
10.
Arterioscler Thromb Vasc Biol ; 23(1): 38-44, 2003 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12524222

RESUMO

OBJECTIVE: Angiotensin II (AII) has been shown to increase endothelial NAD(P)H oxidase activity, which is a source of superoxide anion that in turn may induce the formation of peroxynitrite. Estrogen (E2) has been reported to have vascular protective effects. In this study, we hypothesized that E2 reduces the AII-induced expression of NAD(P)H oxidase and peroxynitrite in endothelial cells. METHODS AND RESULTS: Endothelial cells were cultured and stimulated with AII in the absence or presence of E2. Western blots were used to assess nitric oxide synthase (NOS) and NAD(P)H oxidase expression. Immunofluorescence of nitrotyrosine provided evidence of peroxynitrite formation. Our data indicate that AII increased the expression of endothelial NOS, inducible NOS, and NAD(P)H oxidase in a dose-dependent manner, which was attenuated by incubation with either E2, superoxide dismutase, or the AII type 1 receptor (AT1R) inhibitor candesartan. Estrogen as well as superoxide dismutase also inhibited AII-induced AT(1)R expression and nitrotyrosine staining. The effects of E2 on the AII responses were not inhibited by the E2 receptor antagonist ICI-182,780. CONCLUSIONS: AII stimulation of endothelial cells increases expression of NAD(P)H oxidase and NOS, which may contribute to oxidative stress, as evidenced by peroxynitrite formation. E2 inhibits these AII effects, possibly through reduced AT1R expression.


Assuntos
Angiotensina II/fisiologia , Endotélio Vascular/enzimologia , Estradiol/fisiologia , NADH NADPH Oxirredutases/biossíntese , Óxido Nítrico Sintase/biossíntese , Angiotensina II/antagonistas & inibidores , Angiotensina II/farmacologia , Animais , Capilares/citologia , Capilares/efeitos dos fármacos , Capilares/enzimologia , Bovinos , Linhagem Celular , Vasos Coronários/citologia , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/enzimologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Indução Enzimática/efeitos dos fármacos , Indução Enzimática/fisiologia , Estradiol/farmacologia , NADPH Oxidases , Óxido Nítrico Sintase Tipo II , Ácido Peroxinitroso/biossíntese , Ácido Peroxinitroso/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA