Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Mol Cell Biochem ; 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38478220

RESUMEN

Dietary interventions that modulate the brown adipose tissue (BAT) thermogenic activity could represent a promising therapy for metabolic disorders. In order to examine if dietary walnuts intake regulates the expression of BAT thermogenic markers levels in healthy and metabolically challenged (fructose fed) animals, rats were initially divided into the control and fructose-fed groups. After nine weeks, these groups were subdivided into the one kept on the original regimens and the other supplemented with walnuts. High-fructose diet resulted in an increased relative BAT mass and no change in UCP1 content, while the walnut supplementation increased the amount of UCP1 in BAT, but did not affect 5-HT, NA, DHPG content and DHPG/NA ratio regardless of the diet. Moreover, the CD36 levels were increased following the walnut consumption, unlike FATP1, GLUT1, GLUT4, and glycogen content which remained unchanged. Additionally, the BAT levels of activated IR and Akt were not affected by walnut consumption, while ERK signaling was decreased. Overall, we found that walnut consumption increased UCP1 and CD36 content in the BAT of both control and metabolically challenged rats, suggesting that FFAs represent the BAT preferred substrate under the previously described circumstances. This further implies that incorporating walnuts into the everyday diet may help to alleviate some symptoms of the metabolic disorder.

2.
Int J Mol Sci ; 24(7)2023 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-37047558

RESUMEN

After being discovered over a century ago, insulin was long considered to be a hormone exclusively produced by the pancreas. Insulin presence was later discovered in the brain, which was originally accounted for by its transport across the blood-brain barrier. Considering that both insulin mRNA and insulin were detected in the central nervous system (CNS), it is now known that this hormone is also synthesized in several brain regions, including the hypothalamus, hippocampus, cerebral and cerebellar cortex, and olfactory bulb. Although many roles of insulin in the CNS have been described, it was initially unknown which of them could be attributed to brain-derived and which to pancreatic insulin or whether their actions in the brain overlap. However, more and more studies have been emerging lately, focusing solely on the roles of brain-derived insulin. The aim of this review was to present the latest findings on the roles of brain-derived insulin, including neuroprotection, control of growth hormone secretion, and regulation of appetite and neuronal glucose uptake. Lastly, the impairment of signaling initiated by brain-derived insulin was addressed in regard to memory decline in humans.


Asunto(s)
Sistema Nervioso Central , Insulina , Humanos , Insulina/metabolismo , Sistema Nervioso Central/metabolismo , Encéfalo , Aprendizaje , Barrera Hematoencefálica/metabolismo , Insulina Regular Humana
3.
Int J Mol Sci ; 23(4)2022 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-35216510

RESUMEN

A dysregulated and overwhelming response to an infection accompanied by the exaggerated pro-inflammatory state and metabolism disturbance leads to the fatal outcome in sepsis. Previously we showed that meldonium, an anti-ischemic drug clinically used to treat myocardial and cerebral ischemia, strongly increases mortality in faecal-induced peritonitis (FIP) in rats. We postulated that the same mechanism that is responsible for the otherwise strong anti-inflammatory effects of meldonium could be the culprit of the increased mortality. In the present study, we applied the LPS-induced model of sepsis to explore the presence of any differences from and/or similarities to the FIP model. When it comes to energy production, despite some shared similarities, it is evident that LPS and FIP models of sepsis differ greatly. A different profile of sympathoadrenal activation may account for this observation, as it was lacking in the FIP model, whereas in the LPS model it was strong enough to overcome the effects of meldonium. Therefore, choosing the appropriate model of sepsis induction is of great importance, especially if energy homeostasis is the main focus of the study. Even when differences in the experimental design of the two models are acknowledged, the role of different patterns of energy production cannot be excluded. On that account, our results draw attention to the importance of uninterrupted energy production in sepsis but also call for much-needed revisions of the current recommendations for its treatment.


Asunto(s)
Lipopolisacáridos/farmacología , Metilhidrazinas/farmacología , Sepsis/inducido químicamente , Sepsis/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Inflamación/tratamiento farmacológico , Masculino , Miocardio/patología , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
4.
Int J Mol Sci ; 22(18)2021 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-34575863

RESUMEN

Sepsis is a life-threatening condition caused by the dysregulated and overwhelming response to infection, accompanied by an exaggerated pro-inflammatory state and lipid metabolism disturbance leading to sequential organ failure. Meldonium is an anti-ischemic and anti-inflammatory agent which negatively interferes with lipid metabolism by shifting energy production from fatty acid oxidation to glycolysis, as a less oxygen-demanding pathway. Thus, we investigated the effects of a four-week meldonium pre-treatment on faecal-induced sepsis in Sprague-Dawley male rats. Surprisingly, under septic conditions, meldonium increased animal mortality rate compared with the meldonium non-treated group. However, analysis of the tissue oxidative status did not provide support for the detrimental effects of meldonium, nor did the analysis of the tissue inflammatory status showing anti-inflammatory, anti-apoptotic, and anti-necrotic effects of meldonium. After performing tissue lipidomic analysis, we concluded that the potential cause of the meldonium harmful effect is to be found in the overall decreased lipid metabolism. The present study underlines the importance of uninterrupted energy production in sepsis, closely drawing attention to the possible harmful effects of lipid-mobilization impairment caused by certain therapeutics. This could lead to the much-needed revision of the existing guidelines in the clinical treatment of sepsis while paving the way for discovering new therapeutic approaches.


Asunto(s)
Heces/microbiología , Metilhidrazinas/farmacología , Sepsis/prevención & control , Glándulas Suprarrenales/efectos de los fármacos , Animales , Antiinflamatorios/farmacología , Apoptosis , Biomarcadores , Epinefrina/metabolismo , Ácidos Grasos/metabolismo , Inflamación , Metabolismo de los Lípidos/efectos de los fármacos , Peroxidación de Lípido , Lipidómica , Masculino , Norepinefrina/metabolismo , Estrés Oxidativo , Oxígeno/química , Ratas , Ratas Sprague-Dawley , Temperatura , Resultado del Tratamiento , Triglicéridos/metabolismo , Troponina T/sangre
5.
J Sci Food Agric ; 101(14): 5984-5991, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33856052

RESUMEN

BACKGROUND: Nutritional modulations may be considered a strategy to protect mental health. Neuronal homeostasis is highly dependent on the availability of glucose, which represents the primary energy source for the brain. In this study, we evaluated the effects of walnut intake and fructose-rich diet on the expression of glucose transporters (GLUTs) in two rat brain regions: hypothalamus and hippocampus. RESULTS: Our results show that walnut supplementation of fructose-fed animals restored the hypothalamic content of GLUT1 and GLUT3 protein. Furthermore, walnut intake did not affect increased hypothalamic GLUT2 content upon fructose consumption. These effects were accompanied by distinctive alterations of hippocampal GLUTs levels. Specifically, walnut intake increased GLUT1 content, whereas GLUT2 protein was decreased within the rat hippocampus after both individual and combined treatments. CONCLUSION: Overall, our study suggests that walnut supplementation exerted modulatory effects on the glucose transporters within specific brain regions in the presence of developed metabolic disorder. © 2021 Society of Chemical Industry.


Asunto(s)
Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 2/metabolismo , Transportador de Glucosa de Tipo 3/metabolismo , Hipocampo/metabolismo , Hipotálamo/metabolismo , Juglans/metabolismo , Animales , Fructosa/metabolismo , Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 3/genética , Masculino , Nueces/metabolismo , Ratas , Ratas Wistar
6.
Neurochem Res ; 44(2): 388-399, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30460639

RESUMEN

Our group previously reported that 6-h fasting increased both insulin II mRNA expression and insulin level in rat hypothalamus. Given that insulin effects on central glucose metabolism are insufficiently understood, we wanted to examine if the centrally produced insulin affects expression and/or regional distribution of glucose transporters, and glycogen stores in the hypothalamus during short-term fasting. In addition to determining the amount of total and activated insulin receptor, glucose transporters, and glycogen, we also studied distribution of insulin receptors and glucose transporters within the hypothalamus. We found that short-term fasting did not affect the astrocytic 45 kDa GLUT1 isoform, but it significantly increased the amount of endothelial 55 kDa GLUT1, and neuronal GLUT3 in the membrane fractions of hypothalamic proteins. The level of GLUT2 whose presence was detected in neurons, ependymocytes and tanycytes was also elevated. Unlike hepatic glycogen which was decreased, hypothalamic glycogen content was not changed after 6-h fasting. Our findings suggest that neurons may be given a priority over astrocytes in terms of glucose supply even during the initial phase of metabolic response to fasting. Namely, increase in glucose influx into the brain extracellular fluid and neurons by increasing the translocation of GLUT1, and GLUT3 in the cell membrane may represent the first line of defense in times of scarcity. The absence of co-localization of these membrane transporters with the activated insulin receptor suggests this process takes place in an insulin-independent manner.


Asunto(s)
Astrocitos/metabolismo , Células Endoteliales/metabolismo , Ayuno , Neuronas/metabolismo , Animales , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Hipotálamo/metabolismo , Masculino , Neuroglía/metabolismo , Ratas Wistar , Activación Transcripcional/fisiología
7.
Eur J Neurosci ; 46(1): 1730-1737, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28544147

RESUMEN

In the hypothalamus, insulin takes on many roles involved in energy homoeostasis. Therefore, the aim of this study was to examine hypothalamic insulin expression during the initial phase of the metabolic response to fasting. Hypothalamic insulin content was assessed by both radioimmunoassay and Western blot. The relative expression of insulin mRNA was examined by qPCR. Immunofluorescence and immunohistochemistry were used to determine the distribution of insulin immunopositivity in the hypothalamus. After 6-h fasting, both glucose and insulin levels were decreased in serum but not in the cerebrospinal fluid. Our study showed for the first time that, while the concentration of circulating glucose and insulin decreased, both insulin mRNA expression and insulin content in the hypothalamic parenchyma were increased after short-term fasting. Increased insulin immunopositivity was detected specifically in the neurons of the hypothalamic periventricular nucleus and in the ependymal cells of fasting animals. These novel findings point to the complexity of mechanisms regulating insulin expression in the CNS in general and in the hypothalamus in particular.


Asunto(s)
Ayuno/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Animales , Glucemia/metabolismo , Ayuno/sangre , Ayuno/líquido cefalorraquídeo , Insulina/sangre , Insulina/líquido cefalorraquídeo , Insulina/genética , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar
8.
Biofactors ; 50(1): 101-113, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37482913

RESUMEN

Brown adipose tissue (BAT) converts chemical energy into heat to maintain body temperature. Although fatty acids (FAs) represent a primary substrate for uncoupling protein 1 (UCP1)-dependent thermogenesis, BAT also utilizes glucose for the same purpose. Considering that estrous cycle effects on BAT are not greatly explored, we examined those of 6-h fasting on interscapular BAT (iBAT) thermogenic markers in proestrus and diestrus. We found that the percentage of multilocular adipocytes was lower in proestrus than in diestrus, although it was increased after fasting in both analyzed estrous cycle stages. Furthermore, the percentage of paucilocular adipocytes was increased by fasting, unlike the percentage of unilocular cells, which decreased in both analyzed stages of the estrous cycle. The UCP1 amount was lower in proestrus irrespectively of the examined dietary regimens. Regarding FA transporters, it was shown that iBAT CD36 content was increased in fasted rats in diestrus. In contrast to GLUT1, the level of GLUT4 was interactively modulated by selected estrous cycle phases and fasting. There was no change in insulin receptor and ERK1/2 activation, while AKT activation was interactively modulated by fasting and estrous cycle stages. Our study showed that iBAT exhibits morphological and functional changes in proestrus and diestrus. Moreover, iBAT undergoes additional dynamic functional and morphological changes during short-term fasting to modulate nutrient utilization and adjust energy expenditure.


Asunto(s)
Tejido Adiposo Pardo , Termogénesis , Femenino , Ratas , Animales , Dieta , Ayuno , Ciclo Estral , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
9.
J Exp Biol ; 216(Pt 12): 2302-7, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23580725

RESUMEN

Thermal stressors such as low and high ambient temperature elicit an abundance of neuroendocrine responses including activation of the hypothalamo-pituitary-adrenal (HPA) axis and arginine vasopressin (AVP) release. The exposure to heat is a particularly interesting model for studying AVP action because this kind of stressor represents not only an unpleasant experience but also a threat to osmotic homeostasis. As AVP has long been recognized as a hormone involved in the modulation of HPA axis activity, the aim of this study was to elucidate the role of AVP in acutely heat-exposed rats using Nelivaptan, a selective vasopressin 1b receptor (V1bR) antagonist. Rats were exposed to high ambient temperature (38°C) for 60 min. The circulating hormones were determined by ELISA or chemiluminescence, and intrapituitary adrenocorticotropic hormone (ACTH) and V1bR level were determined by western blot. The results obtained show that V1bR blockade negatively affected the increase in blood ACTH caused by heat exposure. This treatment alone, or in combination with Nelivaptan, decreased intrapituitary V1bR levels while circulating AVP concentration was increased under the same conditions. Furthermore, a strong correlation was observed between blood ACTH and corticosterone concentration. In conclusion, our results directly confirm the positive role of AVP in the regulation of ACTH secretion from the pituitary in animals exposed to heat. Moreover, the results suggest that AVP from the general circulation influences pituitary V1bR.


Asunto(s)
Antagonistas de los Receptores de Hormonas Antidiuréticas , Arginina Vasopresina/sangre , Respuesta al Choque Térmico , Sistema Hipotálamo-Hipofisario/fisiología , Indoles/farmacología , Sistema Hipófiso-Suprarrenal/fisiología , Pirrolidinas/farmacología , Hormona Adrenocorticotrópica/sangre , Hormona Adrenocorticotrópica/metabolismo , Animales , Western Blotting , Corticosterona/sangre , Ensayo de Inmunoadsorción Enzimática , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Mediciones Luminiscentes , Masculino , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Ratas , Ratas Wistar
10.
Acta Biochim Pol ; 69(3): 647-655, 2022 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-35877942

RESUMEN

Appetite regulation in the hypothalamus is dependent on hormonal signals from the periphery, such as insulin and leptin, and can be modulated by both sugar-rich diet and stress. Our aim was to explore the effects of 9-week feeding with 20% fructose solution combined with 4-week chronic unpredictable stress, on appetite-regulating neuropeptides and modulatory role of leptin and insulin signalling in the hypothalamus of male Wistar rats. Energy intake, body mass and adiposity, as well as circulatory leptin and insulin concentrations were assessed. Hypothalamic insulin signalling was analysed at the level of glucose transporters, as well as at the protein level and phosphorylation of insulin receptor, insulin receptor supstrate-1, Akt and ERK. Phosphorylation of AMP-activated protein kinase (AMPK), level of protein tyrosine phosphatase 1B (PTP1B) and expression of leptin receptor (ObRb) and suppressor of cytokine signalling 3 (SOCS3) were also analysed, together with the expression of orexigenic agouti-related protein (AgRP) and anorexigenic proopiomelanocortin (POMC) neuropeptides. The results revealed that stress decreased body mass and adiposity, blood leptin level and expression of ObRb, SOCS3 and POMC, while combination with fructose diet led to marked increase of AgRP, associated with AMPK phosphorylation despite increased plasma insulin. Reduced Akt, enhanced ERK activity and elevated PTP1B were also observed in the hypothalamus of these animals. In conclusion, our results showed that joint effects of fructose diet and stress are more deleterious than the separate ones, since inappropriate appetite control in the hypothalamus may provide a setting for the disturbed energy homeostasis in the long run.


Asunto(s)
Neuropéptidos , Proopiomelanocortina , Proteínas Quinasas Activadas por AMP/metabolismo , Proteína Relacionada con Agouti/metabolismo , Proteína Relacionada con Agouti/farmacología , Animales , Citocinas/metabolismo , Dieta , Fructosa/efectos adversos , Fructosa/metabolismo , Glucosa/metabolismo , Hipotálamo/metabolismo , Insulina , Leptina , Masculino , Neuropéptidos/metabolismo , Neuropéptidos/farmacología , Fosforilación , Proopiomelanocortina/metabolismo , Proopiomelanocortina/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Receptor de Insulina/metabolismo , Receptores de Leptina/metabolismo
11.
Endocrine ; 78(3): 476-483, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36301508

RESUMEN

PURPOSE: Our previous study showed that 6-h fasting increased insulin expression in the hypothalamus of male rats. We, therefore, wanted to examine if this phenomenon occurs in female rats and whether it depended on the estrus cycle phase. METHODS: Female rats in proestrus or diestrus were either exposed to 6-h fasting or had ad libitum access to food. The serum, cerebrospinal fluid, and hypothalamic insulin levels were determined using radioimmunoassay. The hypothalamic insulin mRNA expression was measured by RT-qPCR, while the hypothalamic insulin distribution was assessed immunohistochemically. RESULTS: Albeit the short-term fasting lowered circulating insulin, both hypothalamic insulin mRNA expression and hypothalamic insulin content remained unaltered. As for the hypothalamic insulin distribution, strong insulin immunopositivity was noted primarily in ependymal cells lining the upper part of the third ventricle and some neurons mainly located within the periventricular nucleus. The pattern of insulin distribution was similar between the controls and the females exposed to fasting regardless of the estrous cycle phase. CONCLUSION: The findings of this study indicate that the control of insulin expression in the hypothalamus differs from that in the pancreatic beta cells during short-term fasting. Furthermore, they also imply that the regulation of insulin expression in the female hypothalamus is different from males but independent of the estrus cycle phase.


Asunto(s)
Ayuno , Insulina , Animales , Ratas , Femenino , Masculino , Insulina/metabolismo , Hipotálamo/metabolismo , Estro/fisiología , ARN Mensajero/metabolismo
12.
Arq Bras Cardiol ; 112(1): 67-75, 2019 01.
Artículo en Inglés, Portugués | MEDLINE | ID: mdl-30569948

RESUMEN

BACKGROUND: Prenatal stress may increase risk of developing cardiovascular disorders in adulthood. The cardiotoxic effects of catecholamines are mediated via prolonged adrenergic receptor stimulation and increased oxidative stress upon their degradation by monoamine oxidase A (MAO-A). OBJECTIVES: We investigated long-term effects of prenatal stress on ß (1, 2, 3) adrenergic receptors and MAO-A gene expression in the hearts of adult rat offspring. METHODS: Pregnant rats were exposed to unpredictable mild stress during the third week of gestation. RNA was isolated from left ventricular apex and base of adult offspring. Quantitative PCR was used to measure gene expression in collected ventricular tissue samples. The level of significance was set to p < 0.05. RESULTS: ß3 adrenergic receptor mRNA was undetectable in rat left ventricle. ß1 adrenergic receptor was the predominantly expressed subtype at the apical and basal left ventricular myocardium in the control females. Male offspring from unstressed mothers displayed higher apical cardiac ß1 than ß2 adrenergic receptor mRNA levels. However, ß1 and ß2 adrenergic receptor mRNAs were similarly expressed at the ventricular basal myocardium in males. Unlike males, prenatally stressed females exhibited decreased ß1 adrenergic receptor mRNA expression at the apical myocardium. Prenatal stress did not affect cardiac MAO-A gene expression. CONCLUSIONS: Collectively, our results show that prenatal stress may have exerted region- and sex-specific ß1 and ß2 adrenergic receptor expression patterns within the left ventricle.


Asunto(s)
Monoaminooxidasa/análisis , Miocardio/metabolismo , Preñez/psicología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Receptores Adrenérgicos beta/análisis , Estrés Psicológico/metabolismo , Hormona Adrenocorticotrópica/sangre , Animales , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/psicología , Femenino , Expresión Génica , Ventrículos Cardíacos/metabolismo , Monoaminooxidasa/genética , Madres/psicología , Embarazo , Efectos Tardíos de la Exposición Prenatal/psicología , ARN Mensajero/análisis , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Adrenérgicos beta/genética , Valores de Referencia , Factores Sexuales , Estrés Psicológico/genética , Factores de Tiempo
13.
Acta Histochem ; 121(1): 29-34, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30342863

RESUMEN

It is well known that stress changes levels of pituitary hormones in the bloodstream and in the pituitary itself. However, almost nothing is known about the impact of stress on histological and stereological parameters of the growth hormone producing cells (somatotrophs-GH cells). The aim of the present study was to investigate the effect of: acute and repeated immobilization; acute and repeated restraint on histological and morphofunctional parameters of somatotrophs in adult Wistar rats. Changes in the pituitary gland volume; the volume density and volume of somatotrophs following acute and repeated immobilization (IMO, R-IMO); acute and repeated restraint (R, R-R) were evaluated using a stereological system (newCAST), while growth hormone level within pituitary was determined by Western blot. Our results demonstrated the decrease (p < 0.05) of the pituitary volume (17%, 19%) in the IMO and R groups, respectively, and the increase in the R-R group. The volume density of GH cells decreased (p < 0.05) in the R-IMO (7%), R (26%) and R-R (18%) group in comparison to the control value. The pituitary GH content was increased (p < 0.05) after the IMO (2-fold), R (2.5-fold) and R-R (2.1-fold) as compared to the control group. These results point out that acute and repeated immobilization and/or restraint lead not only to changes in GH hormone concentration, but also modify the morphological aspects of GH cells within the rat pituitary.


Asunto(s)
Hipófisis/patología , Restricción Física , Somatotrofos/patología , Estrés Fisiológico , Animales , Western Blotting , Inmunohistoquímica , Masculino , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT) , Ratas , Ratas Wistar , Estándares de Referencia
14.
Arq. bras. cardiol ; 112(1): 67-75, Jan. 2019. tab, graf
Artículo en Inglés | LILACS | ID: biblio-973833

RESUMEN

Abstract Background: Prenatal stress may increase risk of developing cardiovascular disorders in adulthood. The cardiotoxic effects of catecholamines are mediated via prolonged adrenergic receptor stimulation and increased oxidative stress upon their degradation by monoamine oxidase A (MAO-A). Objectives: We investigated long-term effects of prenatal stress on β (1, 2, 3) adrenergic receptors and MAO-A gene expression in the hearts of adult rat offspring. Methods: Pregnant rats were exposed to unpredictable mild stress during the third week of gestation. RNA was isolated from left ventricular apex and base of adult offspring. Quantitative PCR was used to measure gene expression in collected ventricular tissue samples. The level of significance was set to p < 0.05. Results: β3 adrenergic receptor mRNA was undetectable in rat left ventricle. β1 adrenergic receptor was the predominantly expressed subtype at the apical and basal left ventricular myocardium in the control females. Male offspring from unstressed mothers displayed higher apical cardiac β1 than β2 adrenergic receptor mRNA levels. However, β1 and β2 adrenergic receptor mRNAs were similarly expressed at the ventricular basal myocardium in males. Unlike males, prenatally stressed females exhibited decreased β1 adrenergic receptor mRNA expression at the apical myocardium. Prenatal stress did not affect cardiac MAO-A gene expression. Conclusions: Collectively, our results show that prenatal stress may have exerted region- and sex-specific β1 and β2 adrenergic receptor expression patterns within the left ventricle.


Resumo Fundamento: Estresse pré-natal pode aumentar os riscos de desenvolver doenças cardiovasculares na idade adulta. Os efeitos cardiotóxicos de catecolaminas são mediados pela estimulação prolongada dos receptores adrenérgicos e pelo aumento do estresse oxidativo após sua degradação pela monoamina oxidase A (MAO-A). Objetivos: Investigamos os efeitos a longo prazo de estresse pré-natal nos receptores β (1, 2, 3) adrenérgicos e na expressão do gene MAO-A nos corações da prole adulta de ratos. Método: Ratas prenhes foram expostas a estresse crônico moderado imprevisível durante a terceira semana de gestação. O RNA foi isolado do ápice e da base do ventrículo esquerdo da prole adulta. Utilizou-se PCR quantitativa em tempo real para medir a expressão gênica nas amostras de tecido ventricular coletadas. O nível de significância foi estabelecido em p < 0,05. Resultados: Foi indetectável o mRNA do receptor adrenérgico β3 no ventrículo esquerdo dos ratos. O receptor adrenérgico β1 foi o subtipo mais expresso no miocárdio ventricular esquerdo apical e basal nas fêmeas controle. A prole masculina das mães não estressadas apresentou níveis cardíacos apicais de mRNA do receptor adrenérgico β1 mais altos do que os de β2. Porém, mRNAs dos receptores adrenérgicos β1 e β2 foram expressos de forma semelhante no miocárdio basal ventricular na prole masculina em geral. Ao contrário da prole masculina, a prole feminina exposta ao estresse pré-natal exibiu uma expressão diminuída do mRNA do receptor adrenérgico β1 no miocárdio apical. O estresse pré-natal não afetou a expressão gênica de MAO-A cardíaca. Conclusões: Coletivamente, nossos resultados mostram que estresse pré-natal pode ter exercido padrões de expressão região- e sexo-específica dos receptores adrenérgicos β1 e β2 no ventrículo esquerdo.


Asunto(s)
Animales , Femenino , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Estrés Psicológico/metabolismo , Preñez/psicología , Receptores Adrenérgicos beta/análisis , Monoaminooxidasa/análisis , Miocardio/metabolismo , Efectos Tardíos de la Exposición Prenatal/psicología , Valores de Referencia , Estrés Psicológico/genética , Factores de Tiempo , ARN Mensajero/análisis , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/psicología , Expresión Génica , Factores Sexuales , Receptores Adrenérgicos beta/genética , Ratas Wistar , Hormona Adrenocorticotrópica/sangre , Reacción en Cadena en Tiempo Real de la Polimerasa , Ventrículos Cardíacos/metabolismo , Monoaminooxidasa/genética , Madres/psicología
15.
PLoS One ; 8(5): e63694, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23671692

RESUMEN

Fat mass and obesity associated protein (Fto) is a nucleic acid demethylase, with a preference for thymine or uracil, according to the recent structural data. This fact suggests that methylated single-stranded RNA, rather than DNA, may be the primary Fto substrate. Fto is abundantly expressed in all hypothalamic sites governing feeding behavior. Considering that selective modulation of Fto levels in the hypothalamus can influence food intake, we set out to investigate the effect of 48 h fasting on the Fto expression in lateral hypothalamic area, paraventricular, ventromedial and arcuate nucleus, the regulatory centres of energy homeostasis. We have demonstrated that 48 h fasting causes not only an increase in the overall hypothalamic levels of both Fto mRNA and protein, but also alters Fto intracellular distribution. This switch happens in some neurons of paraventricular and ventromedial nucleus, as well as lateral hypothalamic area, resulting in the majority of the enzyme being localized outside the cell nuclei. Interestingly, the change in the Fto intracellular localization was not observed in neurons of arcuate nucleus, suggesting that fasting did not universally affect Fto in all of the hypothalmic sites involved in energy homeostasis regulation. Both Fto mRNA and catechol-O-methyltransferaze mRNA were upregulated in the identical time-dependent manner in fasting animals. This fact, combined with the knowledge of the Fto substrate preference, may provide further insight into monoamine metabolism in the state of disturbed energy homeostasis.


Asunto(s)
Citoplasma/metabolismo , Hipotálamo/citología , Neuronas/metabolismo , Proteínas/metabolismo , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Animales , Catecol O-Metiltransferasa/metabolismo , Metabolismo Energético , Ayuno/fisiología , Privación de Alimentos , Expresión Génica , Regulación de la Expresión Génica , Homeostasis , Hipotálamo/metabolismo , Masculino , Monoaminooxidasa/genética , Monoaminooxidasa/metabolismo , Transporte de Proteínas , Proteínas/genética , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA