Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Int J Mol Sci ; 23(10)2022 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-35628338

RESUMEN

Low-grade inflammation of the hypothalamus is associated with the disturbance of energy balance. The endocannabinoid system has been implicated in the development and maintenance of obesity as well as in the control of immune responses. The type 2 cannabinoid receptor (CB2) signaling has been associated with anti-inflammatory effects. Therefore, in high fat diet (HFD)-induced obese mice, we modulated CB2 signaling and investigated its effects on energy homeostasis and hypothalamic microgliosis/astrogliosis. We observed no effect on caloric intake and body weight gain in control diet-fed animals that received prolonged icv infusion of the CB2 receptor agonist HU308. Interestingly, we observed a decrease in glucose tolerance in HFD-fed animals treated with HU308. Prolonged icv infusion of HU308 increases astrogliosis in the ventromedial nucleus (VMH) of obese animals and reduced HFD-induced microgliosis in the hypothalamic arcuate (ARC) but not in the paraventricular (PVN) or VMH nuclei. These data indicate that central CB2 signaling modulates glucose homeostasis and glial reactivity in obesogenic conditions, irrespective of changes in body weight.


Asunto(s)
Dieta Alta en Grasa , Gliosis , Animales , Peso Corporal , Encéfalo , Dieta Alta en Grasa/efectos adversos , Glucosa , Hipotálamo , Ratones , Obesidad/etiología
2.
Arch Endocrinol Metab ; 65(5): 549-561, 2021 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-34591411

RESUMEN

OBJECTIVE: Feeding restriction in rats alters the oscillators in suprachiasmatic, paraventricular, and arcuate nuclei, hypothalamic areas involved in food intake. In the present study, using the same animals and experimental protocol, we aimed to analyze if food restriction could reset clock genes (Clock, Bmal1) and genes involved in lipid metabolism (Pgc1a, Pparg, Ucp2) through nutrient-sensing pathways (Sirt1, Ampk, Nampt) in peripheral tissues. METHODS: Rats were grouped according to food access: Control group (CG, food ad libitum), Restricted night-fed (RF-n, food access during 2 h at night), Restricted day-fed (RF-d, food access during 2 h in the daytime), and Day-fed (DF, food access during 12 h in the daytime). After 21 days, rats were decapitated at ZT3 (0900-1000 h), ZT11 (1700-1800 h), or ZT17 (2300-2400 h). Blood, liver, brown (BAT) and peri-epididymal (PAT) adipose tissues were collected. Plasma corticosterone and gene expression were evaluated by radioimmunoassay and qPCR, respectively. RESULTS: In the liver, the expression pattern of Clock and Bmal1 shifted when food access was dissociated from rat nocturnal activity; this phenomenon was attenuated in adipose tissues. Daytime feeding also inverted the profile of energy-sensing and lipid metabolism-related genes in the liver, whereas calorie restriction induced a pre-feeding increased expression of these genes. In adipose tissues, Sirt1 expression was modified by daytime feeding and calorie restriction, with concomitant expression of Pgc1a, Pparg, and Ucp2 but not Ampk and Nampt. CONCLUSION: Feeding restriction reset clock genes and genes involved in lipid metabolism through nutrient-sensing-related genes in rat liver, brown, and peri-epididymal adipose tissues.


Asunto(s)
Hipotálamo , Hígado , Animales , Ritmo Circadiano , Metabolismo de los Lípidos , Hígado/metabolismo , Nutrientes , Ratas
3.
J Neuroendocrinol ; 33(7): e12975, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33942400

RESUMEN

The Wistar audiogenic rat (WAR) strain is used as an animal model of epilepsy, which when submitted to acute acoustic stimulus presents tonic-clonic seizures, mainly dependent on brainstem (mesencephalic) structures. However, when WARs are exposed to chronic acoustic stimuli (audiogenic kindling-AK), they usually present tonic-clonic seizures, followed by limbic seizures, after recruitment of forebrain structures such as the cortex, hippocampus and amygdala. Although some studies have reported that hypothalamic-hypophysis function is also altered in WAR through modulating vasopressin (AVP) and oxytocin (OXT) secretion, the role of these neuropeptides in epilepsy still is controversial. We analyzed the impact of AK and consequent activation of mesencephalic neurocircuits and the recruitment of forebrain limbic (LiR) sites on the hypothalamic-neurohypophysial system and expression of Avpr1a and Oxtr in these structures. At the end of the AK protocol, nine out of 18 WARs presented LiR. Increases in both plasma vasopressin and oxytocin levels were observed in WAR when compared to Wistar rats. These results were correlated with an increase in the expressions of heteronuclear (hn) and messenger (m) RNA for Oxt in the paraventricular nucleus (PVN) in WARs submitted to AK that presented LiR. In the paraventricular nucleus, the hnAvp and mAvp expressions increased in WARs with and without LiR, respectively. There were no significant differences in Avp and Oxt expression in supraoptic nuclei (SON). Also, there was a reduction in the Avpr1a expression in the central nucleus of the amygdala and frontal lobe in the WAR strain. In the inferior colliculus, Avpr1a expression was lower in WARs after AK, especially those without LiR. Our results indicate that both AK and LiR in WARs lead to changes in the hypothalamic-neurohypophysial system and its receptors, providing a new molecular basis to better understaind epilepsy.


Asunto(s)
Epilepsia Refleja , Hipotálamo/metabolismo , Excitación Neurológica/fisiología , Sistemas Neurosecretores/metabolismo , Neurohipófisis/metabolismo , Estimulación Acústica , Animales , Modelos Animales de Enfermedad , Epilepsia Refleja/genética , Epilepsia Refleja/metabolismo , Epilepsia Refleja/patología , Epilepsia Refleja/fisiopatología , Regulación de la Expresión Génica , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/fisiopatología , Hipotálamo/patología , Hipotálamo/fisiopatología , Excitación Neurológica/patología , Masculino , Sistemas Neurosecretores/patología , Sistemas Neurosecretores/fisiopatología , Oxitocina/sangre , Oxitocina/genética , Oxitocina/metabolismo , Neurohipófisis/patología , Neurohipófisis/fisiopatología , Ratas , Ratas Wistar , Convulsiones/genética , Convulsiones/metabolismo , Convulsiones/fisiopatología , Convulsiones/psicología , Vasopresinas/sangre , Vasopresinas/genética , Vasopresinas/metabolismo
4.
Arch. endocrinol. metab. (Online) ; 65(5): 549-561, 2021. tab, graf
Artículo en Inglés | LILACS | ID: biblio-1345196

RESUMEN

ABSTRACT Objective: Feeding restriction in rats alters the oscillators in suprachiasmatic, paraventricular, and arcuate nuclei, hypothalamic areas involved in food intake. In the present study, using the same animals and experimental protocol, we aimed to analyze if food restriction could reset clock genes ( Clock, Bmal1 ) and genes involved in lipid metabolism ( Pgc1a, Pparg, Ucp2 ) through nutrient-sensing pathways ( Sirt1, Ampk, Nampt ) in peripheral tissues. Materials and methods: Rats were grouped according to food access: Control group (CG, food ad libitum ), Restricted night-fed (RF-n, food access during 2 h at night), Restricted day-fed (RF-d, food access during 2 h in the daytime), and Day-fed (DF, food access during 12 h in the daytime). After 21 days, rats were decapitated at ZT3 (0900-1000 h), ZT11 (1700-1800 h), or ZT17 (2300-2400 h). Blood, liver, brown (BAT) and peri-epididymal (PAT) adipose tissues were collected. Plasma corticosterone and gene expression were evaluated by radioimmunoassay and qPCR, respectively. Results: In the liver, the expression pattern of Clock and Bmal1 shifted when food access was dissociated from rat nocturnal activity; this phenomenon was attenuated in adipose tissues. Daytime feeding also inverted the profile of energy-sensing and lipid metabolism-related genes in the liver, whereas calorie restriction induced a pre-feeding increased expression of these genes. In adipose tissues, Sirt1 expression was modified by daytime feeding and calorie restriction, with concomitant expression of Pgc1a , Pparg , and Ucp2 but not Ampk and Nampt . Conclusion: Feeding restriction reset clock genes and genes involved in lipid metabolism through nutrient-sensing-related genes in rat liver, brown, and peri-epididymal adipose tissues.


Asunto(s)
Animales , Ratas , Hipotálamo , Hígado/metabolismo , Nutrientes , Ritmo Circadiano , Metabolismo de los Lípidos
5.
Horm Behav ; 120: 104690, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31954709

RESUMEN

Changes to neonatal nutrition result in long-lasting impairments in energy balance, which may be described as metabolic programing. Astrocytes, which are interconnected by gap junctions, have emerged as important players in the hypothalamic control of food intake. In order to study the effects of nutritional programming on glial morphology and protein expression, cross-fostered male Wistar rats at postnatal day 3 were assigned to three groups based on litter size: small litter (3 pups per dam, SL), normal litter (10 pups per dam, NL), and large litter (16 pups per dam, LL). Rats from the SL group exhibited higher body weight throughout the study and hyperphagia after weaning. LL animals exhibited hyperphagia, high energy efficiency and catch-up of body weight after weaning. Both the SL and LL groups at postnatal day 60 (PN60) exhibited increased levels of plasma leptin, the Lee index (as an index of obesity), adiposity content, immunoreactivity toward T-cell protein tyrosine phosphatase (TCPTP), and glial fibrillary acidic protein (GFAP) in the arcuate nucleus (ARC) of the hypothalamus. Astrocyte morphology was altered in the ARC of SL and LL animals, and this effect occurred in parallel with a reduction in immunoreactivity toward connexin 30 (CX30). The data obtained demonstrate that both neonatal over- and underfeeding promote not only alterations in the metabolic status but also morphological changes in glial cells in parallel with increasing TCPTP and changes in connexin expression.


Asunto(s)
Fenómenos Fisiológicos Nutricionales de los Animales , Conexinas/genética , Gliosis/etiología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Adiposidad/fisiología , Animales , Animales Recién Nacidos , Conexinas/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Gliosis/genética , Gliosis/metabolismo , Hiperfagia/complicaciones , Hiperfagia/genética , Hiperfagia/metabolismo , Hiperfagia/patología , Hipotálamo/metabolismo , Tamaño de la Camada/fisiología , Masculino , Obesidad/complicaciones , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , Embarazo , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Ratas , Ratas Wistar , Factores Sexuales , Factores de Tiempo
6.
Physiol Behav ; 215: 112793, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31874179

RESUMEN

BACKGROUND: Changes in the nutritional supply during the perinatal period can lead to metabolic disturbances and obesity in adulthood. OBJECTIVE: The divergent litter size model was used to investigate the hypothalamic sensitivity to leptin and ghrelin as well as the mechanisms involved in the disruption of food intake and energy expenditure. METHODS: On postnatal day 3 (P3), male Wistar rats were divided into 3 groups: small litter (SL - 3 pups), normal litter (NL - 10 pups), and large litter (LL - 16 pups). Animals at P60 were intraperitoneally treated with leptin (500 µg/Kg), ghrelin (40 µg/Kg), or vehicle (0.9% NaCl) at 5 pm and the following parameters were assessed: food intake and body weight; immunostaining of p-STAT-3 in the hypothalamus; Western Blotting analysis of p-AMPKα and UCP2 in the mediobasal hypothalamus (MBH), and UCP1 in the interscapular brown adipose tissue (BAT); or heat production, VO2, VCO2, and locomotor activity. RESULTS: SL rats had earlier leptin and ghrelin surges, while LL rats had no variations. At P60, after leptin treatment, LL rats showed hypophagia and increased p-STAT-3 expression in the arcuate nucleus, but SL rats had no response. After ghrelin treatment, LL rats did not have the orexigenic response or AMPKα phosphorylation in the MBH, while SL animals, unexpectedly, decreased body weight gain, without changes in food intake, and increased metabolic parameters and UCP1 expression in the BAT. CONCLUSIONS: Changes in the nutritional supply at early stages of life modify leptin and ghrelin responsiveness in adulthood, programming metabolic and central mechanisms, which contribute to overweight and obesity in adulthood.


Asunto(s)
Ghrelina/metabolismo , Hiperfagia/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Desnutrición/metabolismo , Envejecimiento , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Peso Corporal , Ingestión de Alimentos , Metabolismo Energético/fisiología , Femenino , Tamaño de la Camada , Masculino , Obesidad/etiología , Embarazo , Ratas , Ratas Wistar , Factor de Transcripción STAT3/metabolismo
7.
Acta Physiol (Oxf) ; 228(3): e13373, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31483934

RESUMEN

AIM: Whereas some patients have important changes in body core temperature (Tb) during systemic inflammation, others maintain a normal Tb, which is intrinsically associated to immune paralysis. One classical model to study immune paralysis is the use of repeated administration of lipopolysaccharide (LPS), the so-called endotoxin tolerance. However, the neuroimmune mechanisms of endotoxin tolerance remain poorly understood. Hydrogen sulphide (H2 S) is a gaseous neuromodulator produced in the brain by the enzyme cystathionine ß-synthase (CBS). The present study assessed whether endotoxin tolerance is modulated by hypothalamic H2 S. METHODS: Rats with central cannulas (drug microinjection) and intraperitoneal datalogger (temperature record) received a low-dose of lipopolysaccharide (LPS; 100 µg kg-1 ) daily for four consecutive days. Hypothalamic CBS expression and H2 S production rate were assessed, together with febrigenic signalling. Tolerant rats received an inhibitor of H2 S synthesis (AOA, 100 pmol 1 µL-1 icv) or its vehicle in the last day. RESULTS: Antero-ventral preoptic area of the hypothalamus (AVPO) H2 S production rate and CBS expression were increased in endotoxin-tolerant rats. Additionally, hypothalamic H2 S inhibition reversed endotoxin tolerance reestablishing fever, AVPO and plasma PGE2 levels without altering the absent plasma cytokines surges. CONCLUSION: Endotoxin tolerance is not simply a reflection of peripheral reduced cytokines release but actually results from a complex set of mechanisms acting at multiple levels. Hypothalamic H2 S production modulates most of these mechanisms.


Asunto(s)
Dinoprostona/biosíntesis , Endotoxinas/farmacología , Sulfuro de Hidrógeno/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Animales , Cistationina betasintasa/genética , Cistationina betasintasa/metabolismo , Citocinas/metabolismo , Dinoprostona/antagonistas & inhibidores , Dinoprostona/metabolismo , Modelos Animales de Enfermedad , Tolerancia a Medicamentos , Fiebre/tratamiento farmacológico , Fiebre/metabolismo , Lipopolisacáridos/farmacología , Masculino , Área Preóptica/efectos de los fármacos , Área Preóptica/metabolismo , Ratas , Ratas Wistar
8.
J Endocrinol ; 242(2): 125-138, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31189132

RESUMEN

Adrenalectomy (ADX) induces hypophagia and glucocorticoids counter-regulate the peripheral metabolic effects of insulin. This study evaluated the effects of ADX on ICV (lateral ventricle) injection of insulin-induced changes on food intake, mRNA expression of hypothalamic neuropeptides (insulin receptor (InsR), proopiomelanocortin, cocaine and amphetamine-regulated transcript (Cart), agouti-related protein, neuropeptide Y (Npy) in the arcuate nucleus of the hypothalamus (ARC), corticotrophin-releasing factor in the paraventricular nucleus of the hypothalamus) and hypothalamic protein content of insulin signaling-related molecules (insulin receptor substrate (IRS) 1, protein kinase B (AKT), extracellular-signal-regulated kinase (ERK1/2), c-Jun N-terminal kinase (JNK), protein tyrosine phosphatase-1B (PTP1B) and T cell protein tyrosine phosphatase (TCPTP)) Compared with sham animals, ADX increased the hypothalamic content of pJNK/JNK, PTP1B and TCPTP, as well as decreased mRNA expression of InsR, and corticosterone (B) treatment reversed these effects. Insulin central injection enhanced hypothalamic content of pAKT/AKT and Cart mRNA expression, decreased Npy mRNA expression and food intake only in sham rats, without effects in ADX and ADX + B rats. Insulin did not alter the hypothalamic phosphorylation of IRS1 and ERK1/2 in the three experimental groups. These data demonstrate that ADX reduces the expression of InsR and increases insulin counter-regulators in the hypothalamus, as well as ADX abolishes hypophagia, activation of hypothalamic AKT pathway and changes in Cart and Npy mRNA expression in the ARC induced by insulin. Thus, the higher levels of insulin counter-regulatory proteins and lower expression of InsR in the hypothalamus are likely to underlie impaired insulin-induced hypophagia and responses in the hypothalamus after ADX.


Asunto(s)
Adrenalectomía/métodos , Ingestión de Alimentos/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Insulina/farmacología , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Corticosterona/farmacología , Expresión Génica/efectos de los fármacos , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/farmacología , Hipotálamo/metabolismo , Inyecciones Intraventriculares , Insulina/administración & dosificación , Masculino , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Ratas Wistar , Receptor de Insulina/genética , Receptor de Insulina/metabolismo
9.
Mol Cell Endocrinol ; 482: 62-69, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30572001

RESUMEN

Leptin and LPS has been implicated in the development of hypothalamic astrogliosis in rodents. Astrocytes, which are interconnected by gap junction proteins, have emerged as important players in the control of energy homeostasis exerted by the hypothalamus. To investigate the hypothesis of action of T-cell protein tyrosine phosphatase (TCPTP) on the astrocyte morphology, astrocytes from the hypothalamus of one-day-old rats were stimulated with leptin and LPS (used as a positive control). Leptin and LPS induced a marked increase in astrocyte size, an increase in Ptpn2 (TCPTP gene) and gap junction alpha-1 protein, - Gja1 (connexin 43 - CX43 gene) mRNA expression and a decrease in gap junction protein, alpha 6 - Gja6 (CX30 gene) mRNA expression. Remarkably, these effects on astrocytes morphology and connexins were prevented by Ptpn2 siRNA. Astrocytes are known to produce cytokines; here we show that TCPTP acts as an important regulator of the cytokines and it possesses a reciprocal interplay with protein tyrosine phosphatase 1B (PTP1B). Our findings demonstrate that leptin and LPS alter astrocyte morphology by increasing TCPTP, which in turn modulates connexin 30 (CX30) and connexin 43 (CX43) expression. TCPTP and PTP1B seem to act in the regulation of cytokine production in astrocytes.


Asunto(s)
Astrocitos/citología , Hipotálamo/citología , Leptina/efectos adversos , Lipopolisacáridos/efectos adversos , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Células Cultivadas , Conexina 30/genética , Conexina 43 , Citocinas/metabolismo , Hipotálamo/efectos de los fármacos , Tamaño de los Órganos/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Ratas , Ratas Wistar , Regulación hacia Arriba
10.
Int J Mol Sci ; 18(7)2017 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-28677618

RESUMEN

Metabolic endotoxemia contributes to low-grade inflammation in obesity, which causes insulin resistance due to the activation of intracellular proinflammatory pathways, such as the c-Jun N-terminal Kinase (JNK) cascade in the hypothalamus and other tissues. However, it remains unclear whether the proinflammatory process precedes insulin resistance or it appears because of the development of obesity. Hypothalamic low-grade inflammation was induced by prolonged lipopolysaccharide (LPS) exposure to investigate if central insulin resistance is induced by an inflammatory stimulus regardless of obesity. Male Wistar rats were treated with single (1 LPS) or repeated injections (6 LPS) of LPS (100 µg/kg, IP) to evaluate the phosphorylation of the insulin receptor substrate-1 (IRS1), Protein kinase B (AKT), and JNK in the hypothalamus. Single LPS increased the expression of pIRS1, pAKT, and pJNK, whereas the repeated LPS treatment failed to recruit pIRS1 and pAKT. The 6 LPS treated rats showed increased total JNK and pJNK. The 6 LPS rats became unresponsive to the hypophagic effect induced by central insulin administration (12 µM/5 µL, ICV). Prolonged exposure to LPS (24 h) impaired the insulin-induced AKT phosphorylation and the translocation of the transcription factor forkhead box protein O1 (FoxO1) from the nucleus to the cytoplasm of the cultured hypothalamic GT1-7 cells. Central administration of the JNK inhibitor (20 µM/5 µL, ICV) restored the ability of insulin to phosphorylate IRS1 and AKT in 6 LPS rats. The present data suggest that an increased JNK activity in the hypothalamus underlies the development of insulin resistance during prolonged exposure to endotoxins. Our study reveals that weight gain is not mandatory for the development of hypothalamic insulin resistance and the blockade of proinflammatory pathways could be useful for restoring the insulin signaling during prolonged low-grade inflammation as seen in obesity.


Asunto(s)
Peso Corporal , Hipotálamo/metabolismo , Inflamación/etiología , Inflamación/metabolismo , Resistencia a la Insulina , Lipopolisacáridos/efectos adversos , Animales , Modelos Animales de Enfermedad , Endotoxemia , Inflamación/patología , Insulina/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Neuronas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Transducción de Señal
11.
Horm Behav ; 93: 166-174, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28576646

RESUMEN

Acute administration of lipopolysaccharide (LPS) from Gram-negative bacteria induces hypophagia. However, the repeated administration of LPS leads to desensitization of hypophagia, which is associated with increased hypothalamic p-AMPK expression. Because ghrelin and endocannabinoids modulate AMPK activity in the hypothalamus, we hypothesized that these neuromodulators play a role in the reversal of tolerance to hypophagia in rats under long-term exposure to LPS. Male Wistar rats were treated with single (1 LPS, 100µg/kg body weight, ip) or repeated injections of LPS over 6days (6 LPS). Food intake was reduced in the 1 LPS, but not in the 6 LPS group. 6 LPS rats showed an increased serum concentration of acylated ghrelin and reduced ghrelin receptor mRNA expression in the hypothalamus. Ghrelin injection (40µg/kg body weight, ip) increased food intake, body weight gain, p-AMPK hypothalamic expression, neuropeptide Y (NPY) and Agouti related peptide (AgRP) mRNA expression in control animals (Saline). However, in 6 LPS rats, ghrelin did not alter these parameters. Central administration of a CB1R antagonist (AM251, 200ng/µl in 5µl/rat) induced hypophagia in 6 LPS animals, suggesting that the endocannabinoid system contributes to preserved food intake during LPS tolerance. In the presence of AM251, the ability of ghrelin to phosphorylate AMPK in the hypothalamus of 6 LPS group was restored, but not its orexigenic effect. Our data highlight that the orexigenic effects of ghrelin require CB1R signaling downstream of AMPK activation. Moreover, CB1R-mediated pathways contribute to the absence of hypophagia during repeated exposure to endotoxin.


Asunto(s)
Adenilato Quinasa/metabolismo , Ghrelina/metabolismo , Hipotálamo/efectos de los fármacos , Lipopolisacáridos/farmacología , Receptor Cannabinoide CB1/metabolismo , Animales , Endocannabinoides/metabolismo , Hipotálamo/metabolismo , Masculino , Neuropéptido Y/metabolismo , Fosforilación/efectos de los fármacos , Piperidinas/farmacología , Pirazoles/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptores de Ghrelina/metabolismo , Transducción de Señal/efectos de los fármacos
12.
J Endocrinol ; 231(2): 167-180, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27613338

RESUMEN

Water deprivation (WD) induces changes in plasma volume and osmolality, which in turn activate several responses, including thirst, the activation of the renin-angiotensin system (RAS) and vasopressin (AVP) and oxytocin (OT) secretion. These systems seem to be influenced by oestradiol, as evidenced by the expression of its receptor in brain areas that control fluid balance. Thus, we investigated the effects of oestradiol treatment on behavioural and neuroendocrine changes of ovariectomized rats in response to WD. We observed that in response to WD, oestradiol treatment attenuated water intake, plasma osmolality and haematocrit but did not change urinary volume or osmolality. Moreover, oestradiol potentiated WD-induced AVP secretion, but did not alter the plasma OT or angiotensin II (Ang II) concentrations. Immunohistochemical data showed that oestradiol potentiated vasopressinergic neuronal activation in the lateral magnocellular PVN (PaLM) and supraoptic (SON) nuclei but did not induce further changes in Fos expression in the median preoptic nucleus (MnPO) or subfornical organ (SFO) or in oxytocinergic neuronal activation in the SON and PVN of WD rats. Regarding mRNA expression, oestradiol increased OT mRNA expression in the SON and PVN under basal conditions and after WD, but did not induce additional changes in the mRNA expression for AVP in the SON or PVN. It also did not affect the mRNA expression of RAS components in the PVN. In conclusion, our results show that oestradiol acts mainly on the vasopressinergic system in response to WD, potentiating vasopressinergic neuronal activation and AVP secretion without altering AVP mRNA expression.


Asunto(s)
Deshidratación/fisiopatología , Estradiol/uso terapéutico , Estrógenos/uso terapéutico , Neuronas/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Supraóptico/efectos de los fármacos , Desequilibrio Hidroelectrolítico/prevención & control , Animales , Arginina Vasopresina/agonistas , Arginina Vasopresina/análisis , Arginina Vasopresina/metabolismo , Conducta Animal/efectos de los fármacos , Deshidratación/terapia , Ingestión de Líquidos/efectos de los fármacos , Terapia de Reemplazo de Estrógeno , Femenino , Fluidoterapia , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuronas/patología , Ovariectomía/efectos adversos , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/patología , Área Preóptica/efectos de los fármacos , Área Preóptica/metabolismo , Área Preóptica/patología , Ratas Wistar , Órgano Subfornical/efectos de los fármacos , Órgano Subfornical/metabolismo , Órgano Subfornical/patología , Núcleo Supraóptico/metabolismo , Núcleo Supraóptico/patología , Núcleo Vestibular Lateral/efectos de los fármacos , Núcleo Vestibular Lateral/metabolismo , Núcleo Vestibular Lateral/patología , Desequilibrio Hidroelectrolítico/sangre , Desequilibrio Hidroelectrolítico/etiología , Desequilibrio Hidroelectrolítico/fisiopatología
13.
Brain Res ; 1650: 21-30, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27566061

RESUMEN

The time course effects of ovarian steroids on kisspeptin and GnRH/LH systems is not totally clarified. We investigated the temporal relationship among kisspeptin and GnRH mRNA and kisspeptin content in the preoptic area (POA), GnRH content and release in the medial basal hypothalamus (MBH) and plasma LH levels under different steroid treatments. Ovariectomized rats treated with oil (OVOO), oil plus single dose of estradiol (OVOE), oil plus single dose of progesterone (OVOP), estradiol for 3 days plus oil (OVEO) or estradiol for 3 days plus progesterone (OVEP) were hourly decapitated from 10:00 to 17:00 or had the MBH microdialyzed from 09:00 to 19:00. Estradiol and progesterone acutely increased POA kisspeptin content without altering POA kisspeptin mRNA levels. Short-term exposure to both hormones stimulated MBH GnRH content, although no GnRH/LH surges had occurred. Chronic estradiol-treatment increased both kisspeptin mRNA levels and content in the POA, demonstrating that long exposure to estradiol is required to activate the whole kisspeptin synthesis machinery. This was followed by the peak in the GnRH/LH release. In estradiol-primed rats, progesterone further increased POA kisspeptin content, amplified and advanced GnRH/LH surges, with no additional change on POA kisspeptin mRNA. The data show an estradiol-induced temporal association between kisspeptin increase in the POA and GnRH/LH surges. Interestingly, the classic action of progesterone in amplifying and accelerating the GnRH/LH surges seems to occur by a mechanism which involves POA kisspeptin system.


Asunto(s)
Estradiol/metabolismo , Hormona Liberadora de Gonadotropina/efectos de los fármacos , Hormona Liberadora de Gonadotropina/metabolismo , Animales , Estradiol/farmacología , Femenino , Gonadotropinas , Hipotálamo/efectos de los fármacos , Hipotálamo Medio/efectos de los fármacos , Kisspeptinas/efectos de los fármacos , Kisspeptinas/metabolismo , Hormona Luteinizante/sangre , Ovariectomía , Área Preóptica/efectos de los fármacos , Progesterona/metabolismo , Progesterona/farmacología , Ratas , Ratas Wistar , Análisis Espacio-Temporal
14.
Mol Brain ; 9: 1, 2016 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-26739966

RESUMEN

BACKGROUND: Rasd1 is a member of the Ras family of monomeric G proteins that was first identified as a dexamethasone inducible gene in the pituitary corticotroph cell line AtT20. Using microarrays we previously identified increased Rasd1 mRNA expression in the rat supraoptic nucleus (SON) and paraventricular nucleus (PVN) of the hypothalamus in response to increased plasma osmolality provoked by fluid deprivation and salt loading. RASD1 has been shown to inhibit adenylyl cyclase activity in vitro resulting in the inhibition of the cAMP-PKA-CREB signaling pathway. Therefore, we tested the hypothesis that RASD1 may inhibit cAMP stimulated gene expression in the brain. RESULTS: We show that Rasd1 is expressed in vasopressin neurons of the PVN and SON, within which mRNA levels are induced by hyperosmotic cues. Dexamethasone treatment of AtT20 cells decreased forskolin stimulation of c-Fos, Nr4a1 and phosphorylated CREB expression, effects that were mimicked by overexpression of Rasd1, and inhibited by knockdown of Rasd1. These effects were dependent upon isoprenylation, as both farnesyltransferase inhibitor FTI-277 and CAAX box deletion prevented Rasd1 inhibition of cAMP-induced gene expression. Injection of lentiviral vector into rat SON expressing Rasd1 diminished, whereas CAAX mutant increased, cAMP inducible genes in response to osmotic stress. CONCLUSIONS: We have identified two mechanisms of Rasd1 induction in the hypothalamus, one by elevated glucocorticoids in response to stress, and one in response to increased plasma osmolality resulting from osmotic stress. We propose that the abundance of RASD1 in vasopressin expressing neurons, based on its inhibitory actions on CREB phosphorylation, is an important mechanism for controlling the transcriptional responses to stressors in both the PVN and SON. These effects likely occur through modulation of cAMP-PKA-CREB signaling pathway in the brain.


Asunto(s)
Hipotálamo/metabolismo , Neuronas/metabolismo , Proteínas ras/metabolismo , Animales , Línea Celular , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Dexametasona/farmacología , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica/efectos de los fármacos , Glucocorticoides/farmacología , Hipotálamo/efectos de los fármacos , Lentivirus/metabolismo , Masculino , Ratones , Modelos Biológicos , Neuronas/efectos de los fármacos , Presión Osmótica/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Fosforilación/efectos de los fármacos , Neurohipófisis/efectos de los fármacos , Neurohipófisis/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Restricción Física , Estrés Fisiológico/efectos de los fármacos , Núcleo Supraóptico/efectos de los fármacos , Núcleo Supraóptico/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos , Proteínas ras/genética
15.
Am J Physiol Regul Integr Comp Physiol ; 309(11): R1358-68, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26468265

RESUMEN

The present study investigated the type 1 cannabinoid receptor (CB1R) as a potential candidate to mediate the homeostatic responses triggered by 24 h of water deprivation, which constitutes primarily a hydroelectrolytic challenge and also significantly impacts energy homeostasis. The present results demonstrated for the first time that CB1R mRNA expression is increased in the hypothalamus of water-deprived (WD) rats. Furthermore, the administration of ACEA, a CB1R selective agonist, potentiated WD-induced dipsogenic effect, whereas AM251, a CB1R antagonist, attenuated not only water but also salt intake in response to WD. In parallel with the modulation of thirst and salt appetite, we confirmed that CB1Rs are essential for the development of appropriated neuroendocrine responses. Although the administration of ACEA or AM251 did not produce any effects on WD-induced arginine vasopressin (AVP) secretion, oxytocin (OXT) plasma concentrations were significantly decreased in WD rats treated with ACEA. At the genomic level, ACEA significantly decreased AVP and OXT mRNA expression in the hypothalamus of WD rats, whereas AM251 potentiated both basal and WD-induced stimulatory effects on the transcription of AVP and OXT genes. In addition, we showed that water deprivation alone upregulated proopiomelanocortin, Agouti-related peptide, melanin-concentrating hormone, and orexin A mRNA levels in the hypothalamus, and that CB1Rs regulate main central peptidergic pathways controlling food intake, being that most of these effects were also significantly influenced by the hydration status. In conclusion, the present study demonstrated that CB1Rs participate in the homeostatic responses regulating fluid balance and energy homeostasis during water deprivation.


Asunto(s)
Metabolismo Energético , Hipotálamo/metabolismo , Receptor Cannabinoide CB1/metabolismo , Privación de Agua , Equilibrio Hidroelectrolítico , Animales , Regulación del Apetito , Arginina Vasopresina/genética , Arginina Vasopresina/metabolismo , Presión Arterial , Conducta Animal , Agonistas de Receptores de Cannabinoides/farmacología , Antagonistas de Receptores de Cannabinoides/farmacología , Metabolismo Energético/efectos de los fármacos , Conducta Alimentaria , Regulación de la Expresión Génica , Hipotálamo/efectos de los fármacos , Masculino , Modelos Animales , Oxitocina/genética , Oxitocina/metabolismo , ARN Mensajero/metabolismo , Ratas Wistar , Receptor Cannabinoide CB1/efectos de los fármacos , Receptor Cannabinoide CB1/genética , Transducción de Señal , Sodio/sangre , Cloruro de Sodio Dietético/administración & dosificación , Factores de Tiempo , Transcripción Genética , Equilibrio Hidroelectrolítico/efectos de los fármacos
16.
Eur J Pharmacol ; 765: 375-83, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26362752

RESUMEN

Fluoxetine, a selective serotonin reuptake inhibitor (SSRI), has effects beyond its antidepressant properties, altering, e.g., mechanisms involved in blood pressure and vasomotor tone control. Although many studies have addressed the acute impact of fluoxetine on the cardiovascular system, there is a paucity of information on the chronic vascular effects of this SSRI. We tested the hypothesis that chronic fluoxetine treatment enhances the vascular reactivity to vasodilator stimuli by increasing nitric oxide (NO) signaling and activation of potassium (K+) channels. Wistar rats were divided into two groups: (I) vehicle (water for 21 days) or (II) chronic fluoxetine (10 mg/kg/day in the drinking water for 21 days). Fluoxetine treatment increased endothelium-dependent and independent vasorelaxation (analyzed by mesenteric resistance arteries reactivity) as well as constitutive NO synthase (NOS) activity, phosphorylation of eNOS at Serine1177 and NO production, determined by western blot and fluorescence. On the other hand, fluoxetine treatment did not alter vascular expression of neuronal and inducible NOS or guanylyl cyclase (GC). Arteries from fluoxetine-treated rats exhibited increased relaxation to pinacidil. Increased acetylcholine vasorelaxation was abolished by a calcium-activated K+ channel (KCa) blocker, but not by an inhibitor of KATP channels. On the other hand, vascular responses to Bay 41-2272 and 8-bromo-cGMP were similar between the groups. In conclusion, chronic fluoxetine treatment increases endothelium-dependent and independent relaxation of mesenteric resistance arteries by mechanisms that involve increased eNOS activity, NO generation, and KCa channels activation. These effects may contribute to the cardiovascular effects associated with chronic fluoxetine treatment.


Asunto(s)
Fluoxetina/administración & dosificación , Arterias Mesentéricas/metabolismo , Óxido Nítrico/biosíntesis , Canales de Potasio Calcio-Activados/metabolismo , Vasoconstricción/fisiología , Animales , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Masculino , Arterias Mesentéricas/efectos de los fármacos , Óxido Nítrico/agonistas , Técnicas de Cultivo de Órganos , Canales de Potasio Calcio-Activados/agonistas , Ratas , Ratas Wistar , Vasoconstricción/efectos de los fármacos
17.
Am J Physiol Endocrinol Metab ; 308(1): E40-50, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25352433

RESUMEN

Leptin resistance is induced by the feedback inhibitors tyrosine phosphatase-1B (PTP1B) and decreased Src homology 2 domain-containing tyrosine phosphatase-2 (SHP-2) signaling. To investigate the participation of PTP1B and SHP-2 in LPS-induced leptin resistance, we injected repeated (6-LPS) intraperitoneal LPS doses (100 µg/kg ip) for comparison with a single (1-LPS) treatment and evaluated the expression of SHP-2, PTP1B, p-ERK1/2, and p-STAT3 in the hypothalamus of male Wistar rats. The single LPS treatment increased the expression of p-STAT3 and PTP1B but not SHP-2. The repeated LPS treatment reduced SHP-2, increased PTP1B, and did not change p-STAT3. We observed that the PTP1B expression induced by the endotoxin was highly colocalized with leptin receptor cells in the hypothalamus of LepRb-IRES-Cre-tdTomato reporter mice. The single, but not the repeated, LPS treatment decreased the food intake and body weight. Leptin had no stimulatory effect on the hypophagia, body weight loss, or pSTAT3 expression in 6-LPS rats, indicating leptin unresponsiveness. Notably, the PTP1B inhibitor (3.0 nmol/rat in 5 µl icv) restored the LPS-induced hypophagia in 6-LPS rats and restored the ability of leptin to reduce food intake and body weight as well as to phosphorylate STAT3 in the arcuate, paraventricular, and ventromedial nuclei of the hypothalamus. The present data suggest that an increased PTP1B expression in the hypothalamus underlies the development of leptin resistance during repeated exposure to LPS. Our findings contribute to understanding the mechanisms involved in leptin resistance during low-grade inflammation as seen in obesity.


Asunto(s)
Resistencia a Medicamentos , Inflamación/metabolismo , Leptina/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/fisiología , Animales , Resistencia a Medicamentos/efectos de los fármacos , Resistencia a Medicamentos/genética , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Lipopolisacáridos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/genética , Obesidad/metabolismo , Ratas , Ratas Wistar , Receptores de Leptina/genética , Receptores de Leptina/metabolismo
18.
Am J Physiol Regul Integr Comp Physiol ; 306(1): R34-44, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24226027

RESUMEN

Hypophagia induced by inflammation is associated with Janus kinase (JAK)-2/signal transducer and activator of transcription (STAT) 3 signaling pathway, and leptin-mediated hypophagia is also mediated by JAK2-STAT3 pathway. We have previously reported that lipopolysaccharide (LPS) did not reduce food intake in leptin-resistant high-fat diet (HFD) rats but maintained body weight loss. We investigated whether changes in p-STAT3 expression in the hypothalamus and brain stem could account for the desensitization of hypophagia in HFD animals after a low LPS dose (100 µg/kg). Wistar rats fed standard diet (3.95 kcal/g) or HFD (6.3 kcal/g) for 8 wk were assigned into control diet-saline, control diet-LPS, HFD-saline, and HFD-LPS groups. LPS reduced feeding in the control diet but not HFD. This group showed no p-STAT3 expression in the paraventricular nucleus (PVN) and ventromedial hypothalamic nucleus (VMH), but sustained, though lower than control, p-STAT3 in the nucleus of the solitary tract (NTS) and raphe pallidus (RPa). LPS decreased body weight in HFD rats and increased Fos expression in the NTS. LPS increased body temperature, oxygen consumption, and energy expenditure in both control diet and HFD rats, and this response was more pronounced in HFD-LPS group. Brown adipose tissue (BAT) thermogenesis and increased energy expenditure seem to contribute to body weight loss in HFD-LPS. This response might be related with increased brain stem activation. In conclusion, LPS activates STAT3-mediated pathway in the hypothalamus and brain stem, leading to hypophagia, however, LPS effects on food intake, but not body weight loss, are abolished by leptin resistance induced by HFD. The preserved STAT3 phosphorylation in the brain stem suggests that unresponsiveness to LPS on STAT3 activation under HFD might be selective to the hypothalamus.


Asunto(s)
Dieta Alta en Grasa , Hipotálamo/efectos de los fármacos , Lipopolisacáridos/farmacología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Peso Corporal/fisiología , Grasas de la Dieta/metabolismo , Ingestión de Alimentos/fisiología , Hipotálamo/metabolismo , Leptina/metabolismo , Masculino , Ratas , Ratas Wistar , Transducción de Señal/fisiología
19.
Regul Pept ; 182: 12-8, 2013 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-23327999

RESUMEN

It is well established that treatment with rimonabant, a CB1 antagonist, decreases food intake and body weight gain. In part, these responses are mediated by increased activity of hypothalamic neurons related with energy homeostasis. However, food consumption is reversed to basal level during prolonged CB1 antagonist treatment, suggesting tolerance to its anorexigenic effect. This study investigated the effects of acute or prolonged CB1 receptor blockade on the expression of hypothalamic neuropeptides involved with energy homeostasis. Male Wistar rats received vehicle, a single dose or daily doses of rimonabant (10 mg/kg by gavage) over 7 days. Food intake, body weight, CRF and CART immunoreactivity, as well as, mRNA expression of hypothalamic neuropeptides were evaluated. In comparison with vehicle treatment, single dose of rimonabant decreased food intake and body weight. Acute rimonabant treatment also increased Fos-CRF and Fos-CART double labeled neurons in the PVN and Fos immunoreactivity in the ARC. We also observed that acute rimonabant treatment increased CRF, CART and TRH mRNA expression in the PVN, while it decreased POMC and NPY mRNA expression in the ARC with no changes in the CART mRNA expression in this nucleus. There was an increase in CB1 mRNA expression in the PVN of rats that received both acute and prolonged-rimonabant treatment. Interestingly, rats subjected to prolonged rimonabant treatment had no changes in food intake, body weight gain, hypothalamic mRNA expression, Fos expression and CRF and CART neuron activation. These data indicate that tolerance to hypophagic effects of CB1 antagonist, rimonabant, is associated with reversion of hypothalamic neuropeptide gene expression related to regulation of energy homeostasis.


Asunto(s)
Adaptación Fisiológica/efectos de los fármacos , Anorexia/fisiopatología , Antagonistas de Receptores de Cannabinoides/farmacología , Hipotálamo/efectos de los fármacos , Neuropéptidos/genética , Piperidinas/farmacología , Pirazoles/farmacología , Receptor Cannabinoide CB1/antagonistas & inhibidores , Animales , Anorexia/genética , Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , ARN Mensajero/genética , Ratas , Ratas Wistar , Rimonabant
20.
Shock ; 39(1): 104-13, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23247127

RESUMEN

Cholecystokinin (CCK) was first described as a gastrointestinal hormone, but its receptors have been located in cardiac and vascular tissues, as well as in immune cells. Our aims were to investigate the role of CCK on lipopolysaccharide (LPS)-induced hypotension and its ability to modulate previously reported inflammatory mediators, therefore affecting cardiovascular function. To conduct these experiments, rats had their jugular vein cannulated for drug administration, and also, the femoral artery cannulated for mean arterial pressure (MAP) and heart rate records. Endotoxemia induced by LPS from Escherichia coli (1.5 mg/kg; i.v.) stimulated the release of CCK, a progressive drop in MAP, and increase in heart rate. Plasma tumor necrosis factor α (TNF-α), interleukin 10 (IL-10), nitrate, vasopressin, and lactate levels were elevated in the endotoxemic rats. The pretreatment with proglumide (nonselective CCK antagonist; 30 mg/kg; i.p.) aggravated the hypotension and also increased plasma TNF-α and lactate levels. On the other hand, CCK (0.4 µg/kg; i.v.) administered before LPS significantly restored MAP, reduced aortic and hepatic inducible nitric oxide synthase (iNOS) production, and elevated plasma vasopressin and IL-10 concentrations; it did not affect TNF-α. Physiological CCK concentration reduced nitrite and iNOS synthesis by peritoneal macrophages, possibly through a self-regulatory IL-10-dependent mechanism. Together, these data suggest a new role for the peptide CCK in modulating MAP, possibly controlling the inflammatory response, stimulating the anti-inflammatory cytokine, IL-10, and reducing vascular and macrophage iNOS-derived nitric oxide production. Based on these findings, CCK could be used as an adjuvant therapeutic agent to improve cardiovascular function.


Asunto(s)
Colecistoquinina/uso terapéutico , Endotoxemia/tratamiento farmacológico , Hipotensión/prevención & control , Mediadores de Inflamación/sangre , Choque Séptico/tratamiento farmacológico , Animales , Aorta/enzimología , Presión Sanguínea/efectos de los fármacos , Colecistoquinina/antagonistas & inhibidores , Colecistoquinina/sangre , Colecistoquinina/farmacología , Evaluación Preclínica de Medicamentos/métodos , Endotoxemia/sangre , Endotoxemia/fisiopatología , Frecuencia Cardíaca/efectos de los fármacos , Interleucina-10/sangre , Ácido Láctico/sangre , Lipopolisacáridos , Hígado/enzimología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Masculino , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Proglumida/farmacología , Ratas , Ratas Wistar , Choque Séptico/sangre , Choque Séptico/fisiopatología , Factor de Necrosis Tumoral alfa/metabolismo , Vasopresinas/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA