Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Am J Physiol Endocrinol Metab ; 324(4): E314-E329, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36652400

RESUMEN

Kisspeptin and γ-amino butyric acid (GABA), synthesized in the central nervous system, are critical for reproduction. Both are also expressed in peripheral organs/tissues critical to metabolic control (liver/pancreas/adipose). Many kisspeptin neurons coexpress GABAB receptors (GABABR) and GABA controls kisspeptin expression and secretion. We developed a unique mouse lacking GABABR exclusively from kisspeptin cells/neurons (Kiss1-GABAB1KO) to evaluate the impact on metabolism/reproduction. We confirmed selective deletion of GABABR from Kiss1 cells in the anteroventral periventricular nucleus/periventricular nucleus continuum (AVPV/PeN; immunofluorescence and PCR) and arcuate nucleus (ARC), medial amygdala (MeA), pituitary, liver, and testes (PCR). Young Kiss1-GABAB1KO males were fertile, with normal LH and testosterone. Kiss1 expression was similar between genotypes in AVPV/PeN, ARC, MeA, bed nucleus of the stria terminalis (BNST), and peripheral organs (testis, liver, pituitary). Kiss1-GABAB1KO males presented higher fasted glycemia and insulin levels, an impaired response to a glucose overload, reduced insulin sensitivity, and marked insulin resistance. Interestingly, when Kiss1-GABAB1KO males got older (9 mo old) their body weight (BW) increased, in part due to an increase in white adipose tissue (WAT). Old Kiss1-GABAB1KO males showed higher fasted insulin, increased pancreatic insulin content, insulin resistance, and significantly decreased pancreatic kisspeptin levels. In sum, lack of GABABR specifically in Kiss1 cells severely impacts glucose homeostasis in male mice, reinforcing kisspeptin involvement in metabolic regulation. These alterations in glucose homeostasis worsened with aging. We highlight the impact of GABA through GABABR in the regulation of the pancreas kisspeptin system in contrast to liver kisspeptin that was not affected.NEW & NOTEWORTHY We developed a unique mouse lacking GABAB receptors specifically in Kiss1 cells to evaluate the impact on reproduction and metabolism. Knockout males showed a severe impact on glucose homeostasis, which worsened with aging. These results reinforce the proposed kisspeptin involvement in metabolic regulation and highlight the impact of GABA through GABABR in the regulation of the peripheral pancreas kisspeptin system.


Asunto(s)
Resistencia a la Insulina , Insulinas , Ratones , Animales , Masculino , Kisspeptinas/genética , Kisspeptinas/metabolismo , Resistencia a la Insulina/genética , Estradiol/metabolismo , Ratones Noqueados , Reproducción/genética , Homeostasis , Ácido gamma-Aminobutírico/metabolismo
2.
Neuroendocrinology ; 112(10): 998-1026, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34963114

RESUMEN

INTRODUCTION: The kisspeptin gene Kiss1 is expressed in two hypothalamic areas: anteroventral periventricular nucleus/periventricular nucleus (AVPV/PeN) and arcuate nucleus (ARC), and also in gonads. Several pieces of evidence suggests that gamma-amino butyric acid B receptors (GABAB) signaling can regulate Kiss1 expression. Here, we inhibited GABAB signaling from PND2 to PND21 and evaluated the hypothalamic-pituitary-gonadal (HPG) axis. METHODS: BALB/c mice were treated on postnatal days 2-21 (PND2-PND21) with CGP55845 (GABAB antagonist) and evaluated in PND21 and adulthood: gene expression (qPCR) in the hypothalamus and gonads, hormones by radioimmunoassay, gonad histochemistry (H&E), puberty onset, and estrous cycles. RESULTS: At PND21, CGP inhibited Kiss1 and Tac2 and increased Pdyn and Gabbr1 in the ARC of both sexes and decreased Th only in female AVPV/PeN. Serum follicle-stimulating hormone (FSH) and testis weight were decreased in CGP-males, and puberty onset was delayed. In adults, Kiss1, Tac2, Pdyn, Pgr, Cyp19a1, and Gad1 were downregulated, while Gabbr1 was upregulated in the ARC of both sexes. In the AVPV/PeN, Kiss1, Th, Cyp19a1, and Pgr were decreased while Gad1 was increased in CGP-females, whereas Cyp19a1 was increased in CGP-males. Serum FSH was increased in CGP-males while prolactin was increased in CGP-females. Testosterone and progesterone were increased in ovaries from CGP-females, in which Kiss1, Cyp19a1, and Esr1 were downregulated while Hsd3b2 was upregulated, together with increased atretic and decreased ovulatory follicles. Testes from CGP-males showed decreased progesterone, increased Gabbr1, Kiss1, Kiss1r, and Esr2 and decreased Cyp19a1, and clear signs of seminiferous tubules atrophy. CONCLUSION: These results demonstrate that appropriate GABAB signaling during this critical prepubertal period is necessary for the normal development of the HPG axis.


Asunto(s)
Kisspeptinas , Progesterona , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Femenino , Hormona Folículo Estimulante , Antagonistas del GABA , Gónadas , Hipotálamo/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Masculino , Ratones , Progesterona/metabolismo , Prolactina/metabolismo , Receptores de Kisspeptina-1/metabolismo , Maduración Sexual/fisiología , Testosterona/metabolismo , Destete
3.
Am J Physiol Endocrinol Metab ; 318(6): E901-E919, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32286880

RESUMEN

Lack of GABAB receptors in GABAB1 knockout mice decreases neonatal ARC kisspeptin 1 (Kiss1) expression in the arcuate nucleus of the hypothalamus (ARC) in females, which show impaired reproduction as adults. Our aim was to selectively impair GABAB signaling during a short postnatal period to evaluate its impact on the reproductive system. Neonatal male and female mice were injected with the GABAB antagonist CGP 55845 (CGP, 1 mg/kg body wt sc) or saline from postnatal day 2 (PND2) to PND6, three times per day (8 AM, 1 PM, and 6 PM). One group was killed on PND6 for collection of blood samples (hormones by radioimmunoassay), brains for gene expression in the anteroventral periventricular nucleus-periventricular nucleus continuum (AVPV/PeN), and ARC micropunches [quantitative PCR (qPCR)] and gonads for qPCR, hormone contents, and histology. A second group of mice was injected with CGP (1 mg/kg body wt sc) or saline from PND2 to PND6, three times per day (8 AM, 1 PM, and 6 PM), and left to grow to adulthood. We measured body weight during development and parameters of sexual differentiation, puberty onset, and estrous cycles. Adult mice were killed, and trunk blood (hormones), brains for qPCR, and gonads for qPCR and hormone contents were obtained. Our most important findings on PND6 include the CGP-induced decrease in ARC Kiss1 and increase in neurokinin B (Tac2) in both sexes; the decrease in AVPV/PeN tyrosine hydroxylase (Th) only in females; the increase in gonad estradiol content in both sexes; and the increase in primordial follicles and decrease in primary and secondary follicles. Neonatally CGP-treated adults showed decreased ARC Kiss1 and ARC gonadotropin-releasing hormone (Gnrh1) and increased ARC glutamic acid decarboxylase 67 (Gad1) only in males; increased ARC GABAB receptor subunit 1 (Gabbr1) in both sexes; and decreased AVPV/PeN Th only in females. We demonstrate that ARC Kiss1 expression is chronically downregulated in males and that the normal sex difference in AVPV/PeN Th expression is abolished. In conclusion, neonatal GABAergic input through GABAB receptors shapes gene expression of factors critical to reproduction.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Hipotálamo Anterior/metabolismo , Receptores de GABA-B/metabolismo , Animales , Animales Recién Nacidos , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Estradiol/metabolismo , Femenino , Hormona Folículo Estimulante/metabolismo , Antagonistas de Receptores de GABA-B/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo Anterior/efectos de los fármacos , Kisspeptinas/genética , Kisspeptinas/metabolismo , Hormona Luteinizante/metabolismo , Masculino , Ratones , Ovario/efectos de los fármacos , Ovario/metabolismo , Ácidos Fosfínicos/farmacología , Propanolaminas/farmacología , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Pubertad/efectos de los fármacos , Pubertad/genética , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Receptores de GABA-B/genética , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Reproducción/efectos de los fármacos , Reproducción/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Diferenciación Sexual/efectos de los fármacos , Diferenciación Sexual/genética , Taquicininas/genética , Taquicininas/metabolismo , Testículo/efectos de los fármacos , Testículo/metabolismo , Testosterona/metabolismo , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
4.
Gen Comp Endocrinol ; 296: 113518, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32474048

RESUMEN

In addition to key mammotrophic hormones such as the pituitary prolactin (PRL) and the ovarian steroids progesterone and estradiol, there are local factors that modulate the tissue dynamics of the mammary glands during pregnancy and lactation. By immunohistochemistry and RT-PCR, we found local transcription and translation of gonadotropin-releasing hormone (GNRH), GNRH receptor (GNRHR), PRL and PRL receptor (PRLR) in mammary glands of adult vizcachas during pregnancy and lactation. Both GNRH and GNRHR showed a lag between protein expression and gene transcription throughout the gestational period: while the highest transcription levels of these genes were recorded at early-pregnancy, the epithelial immunoexpressions of both showed their maximum during lactation. RIA results corroborated the presence of GNRH in mammary glands at all the analyzed stages and confirmed the maximum amount of this peptide in the lactating group. Significant amounts of GNRH were detected in milk samples as well. Conversely, PRL and PRLR shared similar protein and gene expression profiles, all exhibiting maximum values during lactation. GNRH peptide content in mammary glands of females with sulpiride-induced hyperprolactinemia (HP) was significantly lower than that of control females (CT). Although PRL mRNA levels remained unchanged, there was a marked increase in theα-lactalbumin (LALBA) transcription in mammary glands of HP- vs CT-females. These results suggest that after targeting mammary glands, PRL stimulates the expression of milk protein genes, but also, tempers the local expression of GNRH. Mammary gland-explantssupplemented with a GNRH analogue (GN-explants) had no differences in terms of PRLR orLALBA transcription levels compared to CT-explants, so the mammary PRLR signaling would not appear to be modulated by GNRH. Yet, mRNA expression levels of both GNRH and the GNRHR-downstream factor, EGR1, were significantly higher in GN-explants compared to that of CT which would point to a GNRH-positive feedback mechanism. In summary, the local coupled expression of GNRH, GNRHR and EGR1 in the mammary gland throughout pregnancy of vizcachas, the PRL-dependent mammary GNRH secretion as well as the GNRH positive feedback on its own transcription suggest an autocrine-paracrine regulatory mechanism and propose an active role for GNRH in mammary gland tissue remodeling.


Asunto(s)
Regulación de la Expresión Génica , Hormona Liberadora de Gonadotropina/genética , Homeostasis , Glándulas Mamarias Animales/metabolismo , Receptores LHRH/genética , Roedores/genética , Animales , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Epitelio/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/metabolismo , Lactancia/fisiología , Ligandos , Especificidad de Órganos , Embarazo , Prolactina/genética , Prolactina/metabolismo , ARN Mensajero/metabolismo , Receptores LHRH/metabolismo , Receptores de Prolactina/genética , Receptores de Prolactina/metabolismo , Reproducción , Transducción de Señal/efectos de los fármacos
5.
Reproduction ; 157(6): R225-R233, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30844750

RESUMEN

Neuroendocrine control of reproduction involves the interplay of various factors that become active at some point along development. GnRH is the main neurohormone controlling reproduction and among the most important inputs modulating GnRH synthesis/secretion are GABA and kisspeptins. These interactions of GABA and kisspeptin in the control of GnRH secretion can take place by the presence of the receptors of both factors on the GnRH neuron or alternatively by the actions of GABA on kisspeptin neurons and/or the actions of kisspeptin on GABA neurons. Kisspeptin acts on the Kiss1R, a seven transmembrane domain, Gαq/11-coupled receptor that activates phospholipase C, although some Gαq/11-independent pathways in mediating part of the effects of Kiss1R activation have also been proposed. GABA acts through two kinds of receptors, ionotropic GABAA/C receptors involving a chloride channel and associated with fast inhibitory/stimulatory conductance and metabotropic GABAB receptors (GABABR) that are Gi/0 protein linked inducing late slow hyperpolarization. In this review, we aim to summarize the different ways in which these two actors, kisspeptin and GABA, interact to modulate GnRH secretion across the reproductive lifespan.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Receptores de Kisspeptina-1/metabolismo , Reproducción/fisiología , Ácido gamma-Aminobutírico/metabolismo , Animales , Humanos
6.
Gen Comp Endocrinol ; 273: 40-51, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29656043

RESUMEN

The South American plains vizcacha, Lagostomus maximus, is a caviomorph rodent native from Argentina, Bolivia and Paraguay. It shows peculiar reproductive features like pre-ovulatory follicle recruitment during pregnancy with an ovulatory process at around mid-gestation. We have described the activation of the hypothalamic - pituitary - ovarian (HPO) axis during pregnancy. A progressive decrease of progesterone (P4) at mid-pregnancy elicits the delivery of gonadotropin-releasing hormone (GnRH) with the consequent secretion of follicle stimulating hormone (FSH) and estradiol (E2) followed by luteinizing hormone (LH) release resulting in follicular luteinization and the P4 concentration recover. Pituitary gland is the central regulator of the HPO axis being E2 a key hormone involved in the regulation of its activity. In this work we analyzed the action of E2 on the pituitary response to the GnRH wave as well as its involvement on LH secretion at mid-gestation in L. maximus. The expression of GnRHR at the pituitary pars distalis showed a significant decrease at mid-pregnancy compared to early- and term-gestating females. ERα showed a significant increment from mid-gestation whereas ERß did not show variations throughout pregnancy; whereas the LH expression in the pituitary pars distalis showed a significant increase at mid-gestation, concordantly with serum LH, which was followed by a decrease at term-gestation with similar values than at early-pregnancy. The number of cells with co-localization of ERα and GnRHR showed a decline at mid-pregnancy related to early- and term-gestation, whereas the cells with co-localization of ERα and LH increased at mid- and term-pregnancy. On the other hand, ex vivo measuring of LH pulsatility showed a significant increment in the total mass of LH delivered at mid-pregnancy followed by a decrease at term-gestation. The stimulation of ERα with the PPT specific agonist induced a significant increment in the total mass of LH released, whereas no changes were determined when ERß was stimulated with its specific agonist MPP. These results suggest that LH pulsatility rise at mid-pregnancy would be enabled by the increase of E2 acting through ERα.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Hormona Luteinizante/metabolismo , Hipófisis/metabolismo , Roedores/metabolismo , Animales , Antineoplásicos Hormonales , Receptor beta de Estrógeno/metabolismo , Femenino , Adenohipófisis/metabolismo , Embarazo , Receptores LHRH/metabolismo
7.
Am J Physiol Endocrinol Metab ; 311(2): E380-95, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27329801

RESUMEN

Type 1 diabetes (T1D) originates from autoimmune ß-cell destruction. IMT504 is an immunomodulatory oligonucleotide that increases mesenchymal stem cell cloning capacity and reverts toxic diabetes in rats. Here, we evaluated long-term (20 doses) and short-term (2-6 doses) effects of IMT504 (20 mg·kg(-1)·day(-1) sc) in an immunodependent diabetes model: multiple low-dose streptozotocin-injected BALB/c mice (40 mg·kg(-1)·day(-1) ip for 5 consecutive days). We determined blood glucose, glucose tolerance, serum insulin, islet morphology, islet infiltration, serum cytokines, progenitor cell markers, immunomodulatory proteins, proliferation, apoptosis, and islet gene expression. IMT504 reduced glycemia, induced ß-cell recovery, and impaired islet infiltration. IMT504 induced early blood glucose decrease and infiltration inhibition, increased ß-cell proliferation and decreased apoptosis, increased islet indoleamine 2,3-dioxygenase (IDO) expression, and increased serum tumor necrosis factor and interleukin-6 (IL-6). IMT504 affected islet gene expression; preproinsulin-2, proglucagon, somatostatin, nestin, regenerating gene-1, and C-X-C motif ligand-1 cytokine (Cxcl1) increased in islets from diabetic mice and were decreased by IMT504. IMT504 downregulated platelet endothelial cell adhesion molecule-1 (Pecam1) in islets from control and diabetic mice, whereas it increased regenerating gene-2 (Reg2) in islets of diabetic mice. The IMT504-induced increase in IL-6 and islet IDO expression and decreased islet Pecam1 and Cxcl1 mRNA expression could participate in keeping leukocyte infiltration at bay, whereas upregulation of Reg2 may mediate ß-cell regeneration. We conclude that IMT504 effectively reversed immunodependent diabetes in mice. Corroboration of these effects in a model of autoimmune diabetes more similar to human T1D could provide promising results for the treatment of this disease.


Asunto(s)
Glucemia/efectos de los fármacos , Citocinas/efectos de los fármacos , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Oligodesoxirribonucleótidos/farmacología , ARN Mensajero/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Glucemia/metabolismo , Proliferación Celular/efectos de los fármacos , Quimiocina CXCL1/efectos de los fármacos , Quimiocina CXCL1/genética , Citocinas/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 1/genética , Modelos Animales de Enfermedad , Prueba de Tolerancia a la Glucosa , Indolamina-Pirrol 2,3,-Dioxigenasa/efectos de los fármacos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Interleucina-6/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Litostatina/efectos de los fármacos , Litostatina/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Nestina/efectos de los fármacos , Nestina/genética , Proteínas Asociadas a Pancreatitis , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/efectos de los fármacos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Proglucagón/efectos de los fármacos , Proglucagón/genética , Precursores de Proteínas/efectos de los fármacos , Precursores de Proteínas/genética , Proteínas/efectos de los fármacos , Proteínas/genética , ARN Mensajero/metabolismo , Somatostatina/efectos de los fármacos , Somatostatina/genética , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Transcriptoma/efectos de los fármacos , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
8.
Gen Comp Endocrinol ; 232: 174-84, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-26704854

RESUMEN

Gonadotropin-releasing hormone (GnRH) is the regulator of the hypothalamic-hypophyseal-gonadal (HHG) axis. GnRH and GAP (GnRH-associated protein) are both encoded by a single preprohormone. Different variants of GnRH have been described. In most mammals, GnRH is secreted in a pulsatile manner that stimulates the release of follicle-stimulating hormone (FSH) and luteinizing hormone (LH). The South-American plains vizcacha, Lagostomus maximus, is a rodent with peculiar reproductive features including natural poly-ovulation up to 800 oocytes per estrous cycle, pre-ovulatory follicle formation throughout pregnancy and an ovulatory process which takes place at mid-gestation and adds a considerable number of secondary corpora lutea. Such features should occur under a special modulation of the HHG axis, guided by GnRH. The aim of this study was to sequence hypothalamic GnRH preprogonadotrophin mRNA in the vizcacha, to compare it with evolutionarily related species and to identify its expression, distribution and pulsatile pattern of secretion. The GnRH1variant was detected and showed the highest homology with that of chinchilla, its closest evolutionarily related species. Two isoforms of transcripts were identified, carrying the same coding sequence, but different 5' untranslated regions. This suggests a sensitive equilibrium between RNA stability and translational efficiency. A predominant hypothalamic localization and a pulsatile secretion pattern of one pulse of GnRH every hour were found. The lower homology found for GAP, also among evolutionarily related species, depicts a potentially different bioactivity.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Animales , Femenino , Embarazo , Análisis de Secuencia , América del Sur , Distribución Tisular
9.
Am J Physiol Endocrinol Metab ; 304(10): E1064-76, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23531612

RESUMEN

Adult mice lacking functional GABAB receptors (GABAB1KO) have glucose metabolism alterations. Since GABAB receptors (GABABRs) are expressed in progenitor cells, we evaluated islet development in GABAB1KO mice. Postnatal day 4 (PND4) and adult, male and female, GABAB1KO, and wild-type littermates (WT) were weighed and euthanized, and serum insulin and glucagon was measured. Pancreatic glucagon and insulin content were assessed, and pancreas insulin, glucagon, PCNA, and GAD65/67 were determined by immunohistochemistry. RNA from PND4 pancreata and adult isolated islets was obtained, and Ins1, Ins2, Gcg, Sst, Ppy, Nes, Pdx1, and Gad1 transcription levels were determined by quantitative PCR. The main results were as follows: 1) insulin content was increased in PND4 GABAB1KO females and in both sexes in adult GABAB1KOs; 2) GABAB1KO females had more clusters (<500 µm(2)) and less islets than WT females; 3) cluster proliferation was decreased at PND4 and increased in adult GABAB1KO mice; 4) increased ß-area at the expense of the α-cell area was present in GABAB1KO islets; 5) Ins2, Sst, and Ppy transcription were decreased in PND4 GABAB1KO pancreata, adult GABAB1KO female islets showed increased Ins1, Ins2, and Sst expression, Pdx1 was increased in male and female GABAB1KO islets; and 6) GAD65/67 was increased in adult GABAB1KO pancreata. We demonstrate that several islet parameters are altered in GABAB1KO mice, further pinpointing the importance of GABABRs in islet physiology. Some changes persist from neonatal ages to adulthood (e.g., insulin content in GABAB1KO females), whereas other features are differentially regulated according to age (e.g., Ins2 was reduced in PND4, whereas it was upregulated in adult GABAB1KO females).


Asunto(s)
Resistencia a la Insulina/fisiología , Islotes Pancreáticos/fisiología , Receptores de GABA-B/deficiencia , Animales , Animales Recién Nacidos , Peso Corporal/fisiología , Femenino , Regulación de la Expresión Génica , Glucagón/sangre , Glucagón/genética , Glucagón/fisiología , Glutamato Descarboxilasa/fisiología , Insulina/sangre , Insulina/genética , Insulina/fisiología , Islotes Pancreáticos/crecimiento & desarrollo , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/ultraestructura , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Tamaño de los Órganos/fisiología , Antígeno Nuclear de Célula en Proliferación/análisis , Antígeno Nuclear de Célula en Proliferación/fisiología , ARN/química , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
10.
Biol Reprod ; 89(5): 115, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24089203

RESUMEN

In mammals, elevated levels of progesterone (P4) throughout gestation maintain a negative feedback over the hypothalamic-hypophyseal-gonadal (H-H-G) axis, avoiding preovulatory follicular growth and preventing ovulation. Recent studies showed that in the South American plains vizcacha (Lagostomus maximus) folliculogenesis progresses to preovulatory stages during gestation, and an ovulatory process seems to occur at midgestation. The aim of this work was to analyze hypothalamic gonadotropin-releasing hormone (GnRH) and P4 receptors (PR) expression and luteinizing hormone (LH) secretion and correlate these with the functional state of the ovary in nonovulating and ovulating females and gestating females with special emphasis in the supposedly ovulating females at midgestation. We investigated P4 and LH serum levels as well as the distribution, localization, and expression of PR and GnRH in the hypothalamus of L. maximus at different time points during gestation and in nongestating, ovulating and nonovulating, females. A significant increment in GnRH, P4, and LH was detected in midpregnant vizcachas with respect to early-pregnant and to ovulating females. PR was also significantly increased in midpregnant animals. PR was detected in neurons of the preoptic and hypothalamic areas. Coexistence of both PR and GnRH in neurons of medial preoptic area and supraoptic nucleus was detected. Midpregnant animals showed increased number of PR immunoreactive cells at median eminence, localized adjacently to GnRH immunoreactive fibers. High expression of hypothalamic GnRH and PR, despite an increased level of P4, was correlated with the presence of antral, preovulatory follicles, and luteinized unruptured follicles at midgestation that suggest a possible role of the H-H-G axis in the modulation of ovulation during gestation in L. maximus.


Asunto(s)
Hormona Liberadora de Gonadotropina/genética , Hipotálamo/metabolismo , Preñez , Receptores de Progesterona/genética , Roedores/genética , Animales , Femenino , Edad Gestacional , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Luteinizante/genética , Hormona Luteinizante/metabolismo , Ovulación/fisiología , Embarazo , Preñez/genética , Preñez/metabolismo , Receptores de Progesterona/metabolismo , Roedores/metabolismo , América del Sur
11.
Reproduction ; 145(6): 609-20, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23613616

RESUMEN

Dietary protein restriction during pregnancy and lactation in rats impairs ß-cell function and mass in neonates and leads to glucose intolerance in adult offspring. Maternal taurine (Tau) supplementation during pregnancy in rats restores ß-cell function and mass in neonates, but its long-term effects are unclear. The prevention of postnatal catch-up growth has been suggested to improve glucose tolerance in adult offspring of low-protein (LP)-fed mothers. The objective of this study was to examine the relative contribution of ß-cell dysfunction and insulin resistance to impaired glucose tolerance in 130-day-old rat offspring of LP-fed mothers and the effects of maternal Tau supplementation on ß-cell function and insulin resistance in these offspring. Pregnant rats were fed i) control, ii) LP, and iii) LP+Tau diets during gestation and lactation. Offspring were given a control diet following weaning. A fourth group consisting of offspring of LP-fed mothers, maintained on a LP diet following weaning, was also studied (LP-all life). Insulin sensitivity in the offspring of LP-fed mothers was reduced in females but not in males. In both genders, LP exposure decreased ß-cell function. Tau supplementation improved insulin sensitivity in females and ß-cell function in males. The LP-all life diet improved ß-cell function in males. We conclude that i) maternal Tau supplementation has persistent effects on improving glucose metabolism (ß-cell function and insulin sensitivity) in adult rat offspring of LP-fed mothers and ii) increasing the amount of protein in the diet of offspring adapted to a LP diet after weaning may impair glucose metabolism (ß-cell function) in a gender-specific manner.


Asunto(s)
Diabetes Mellitus Tipo 2/prevención & control , Dieta con Restricción de Proteínas/efectos adversos , Suplementos Dietéticos , Resistencia a la Insulina , Células Secretoras de Insulina/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos , Taurina/uso terapéutico , Animales , Péptido C/sangre , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/patología , Proteínas en la Dieta/efectos adversos , Femenino , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/patología , Lactancia , Masculino , Embarazo , Complicaciones del Embarazo/etiología , Complicaciones del Embarazo/fisiopatología , Deficiencia de Proteína/etiología , Deficiencia de Proteína/fisiopatología , Ratas , Ratas Wistar , Caracteres Sexuales , Destete
12.
Neuroendocrinology ; 98(3): 212-23, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24080944

RESUMEN

BACKGROUND/AIMS: Adult mice lacking functional GABAB receptors (GABAB1KO) show altered Gnrh1 and Gad1 expressions in the preoptic area-anterior hypothalamus (POA-AH) and females display disruption of cyclicity and fertility. Here we addressed whether sexual differentiation of the brain and the proper wiring of the GnRH and kisspeptin systems were already disturbed in postnatal day 4 (PND4) GABAB1KO mice. METHODS: PND4 wild-type (WT) and GABAB1KO mice of both sexes were sacrificed; tissues were collected to determine mRNA expression (qPCR), amino acids (HPLC), and hormones (RIA and/or IHC). RESULTS: GnRH neuron number (IHC) did not differ among groups in olfactory bulbs or OVLT-POA. Gnrh1 mRNA (qPCR) in POA-AH was similar among groups. Gnrh1 mRNA in medial basal hypothalamus (MBH) was similar in WTs but was increased in GABAB1KO females compared to GABAB1KO males. Hypothalamic GnRH (RIA) was sexually different in WTs (males > females), but this sex difference was lost in GABAB1KOs; the same pattern was observed when analyzing only the MBH, but not in the POA-AH. Arcuate nucleus Kiss1 mRNA (micropunch-qPCR) was higher in WT females than in WT males and GABAB1KO females. Gad1 mRNA in MBH was increased in GABAB1KO females compared to GABAB1KO males. Serum LH and gonadal estradiol content were also increased in GABAB1KOs. CONCLUSION: We demonstrate that GABABRs participate in the sexual differentiation of the ARC/MBH, because sex differences in several reproductive genes, such as Gad1, Kiss1 and Gnrh1, are critically disturbed in GABAB1KO mice at PND4, probably altering the organization and development of neural circuits governing the reproductive axis.


Asunto(s)
Glutamato Descarboxilasa/deficiencia , Hormona Liberadora de Gonadotropina/deficiencia , Hipotálamo Medio/metabolismo , Kisspeptinas/deficiencia , Precursores de Proteínas/deficiencia , Receptores de GABA-B/deficiencia , Diferenciación Sexual/genética , Animales , Animales Recién Nacidos , Núcleo Arqueado del Hipotálamo/crecimiento & desarrollo , Núcleo Arqueado del Hipotálamo/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Glutamato Descarboxilasa/genética , Hormona Liberadora de Gonadotropina/genética , Hipotálamo Medio/crecimiento & desarrollo , Kisspeptinas/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Precursores de Proteínas/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de GABA-B/genética
13.
Physiol Rep ; 11(15): e15790, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37568265

RESUMEN

We have demonstrated that oligodeoxynucleotide IMT504 promotes significant improvement in the diabetic condition in diverse animal models. Based on these results, here we evaluated whether these effects observed in vivo could be due to direct effects on ß-cells. We demonstrate by immunofluorescence that IMT504 enters the cell and locates in cytoplasm where it induces GSK-3ß phosphorylation that inactivates this kinase. As GSK-3ß tags Pdx1 for proteasomal degradation, by inactivating GSK-3ß, IMT504 induces an increase in Pdx1 protein levels, demonstrated by Western blotting. Concomitantly, an increase in Ins2 and Pdx1 gene transcription was observed, with no significant increase in insulin content or secretion. Enhanced Pdx1 is promising since it is a key transcription factor for insulin synthesis and is also described as an essential factor for the maintenance ß-cell phenotype and function. Dose-dependent inhibition of H2 O2 -induced apoptosis determined by ELISA as well as decreased expression of Bax was also observed. These results were confirmed in another ß-cell line, beta-TC-6 cells, in which a cytokine mix induced apoptosis that was reversed by IMT504. In addition, an inhibitor of IMT504 entrance into cells abrogated the effect IMT504. Based on these results we conclude that the ß-cell recovery observed in vivo may include direct effects of IMT504 on ß-cells, by maintaining their identity/phenotype and protecting them from oxidative stress and cytokine-induced apoptosis. Thus, this work positions IMT504 as a promising option in the framework of the search of new therapies for type I diabetes treatment.


Asunto(s)
Apoptosis , Oligodesoxirribonucleótidos , Animales , Glucógeno Sintasa Quinasa 3 beta , Oligodesoxirribonucleótidos/farmacología , Insulina/metabolismo , Citocinas/farmacología , Proliferación Celular
14.
J Comp Neurol ; 531(7): 720-742, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36716283

RESUMEN

In mammals, gestation is considered a physiological hyperprolactinemia status. Prolactin (PRL) is one of the modulators of gonadotropin-releasing hormone (GnRH) neurons function. The South American plains vizcacha (Lagostomus maximus) is a unique model to study the regulation of hypothalamic GnRH neurons by direct and indirect steroid-dependent pathways. The aim was to characterize the hypothalamic expression of endocrine markers in vizcacha during gestation as well as their response to experimental induced hyperprolactinemia. The possible involvement of PRL regulatory pathways on GnRH in the context of hypothalamic and pituitary reactivation in mid-gestating vizcachas was discussed. Using two in vivo approaches, we determined changes in the hypothalamic expression and distribution of prolactin receptor (PRLR), tyrosine hydroxylase (TH), and dopamine type 2 receptor. A significant increment in the number of tuberoinfundibular dopaminergic (TIDA) neurons was determined in the arcuate nucleus from early to term pregnancy. On the other hand, at preoptic area, the number of both TH+PRLR+ and GnRH+PRLR+ double-labeled neurons significantly decreased at mid-pregnancy probably allowing the recovery of GnRH expression indicating that both types of neurons may represent the key points of PRL indirect and direct pathways modulating GnRH. Moreover, in a model of induced hyperprolactinemic vizcachas, the inhibitory effect of PRL on GnRH at both expression and delivery levels were confirmed. These results suggest the concomitant participation of both PRL regulatory pathways on GnRH modulation and pinpoint the key role of PRL on GnRH expression enabling the recovery of the hypothalamic activity during the gestation in this species.


Asunto(s)
Hormona Liberadora de Gonadotropina , Hiperprolactinemia , Embarazo , Femenino , Animales , Hormona Liberadora de Gonadotropina/metabolismo , Receptores de Prolactina/metabolismo , Hormonas Liberadoras de Hormona Hipofisaria/metabolismo , Hormonas Liberadoras de Hormona Hipofisaria/farmacología , Hiperprolactinemia/metabolismo , Hipotálamo/metabolismo , Roedores/metabolismo , Neuronas Dopaminérgicas/metabolismo
15.
J Comp Physiol B ; 192(1): 141-159, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34459966

RESUMEN

Melatonin, the key messenger of photoperiodic information, is synthesized in the pineal gland by arylalkylamine N-acetyltransferase enzyme (AANAT). It binds to specific receptors MT1 and MT2 located in the hypothalamus and pituitary gland. Melatonin can modulate the reproductive axis affecting the secretion of gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH). The South American plains vizcacha, Lagostomus maximus, shows natural poliovulation of up to 800 oocytes per estrous cycle, a 154-day long pregnancy, and reactivation of the reproductive axis at mid-gestation with pre-ovulatory follicular recruitment, presence of active corpora lutea, and variations of the endocrine status. Here we analyzed the involvement of melatonin in the modulation of the hypothalamic and pituitary gland physiology of vizcacha thorough several approaches, including histological localization of melatoninergic system components, assessment of melatoninergic components expression throughout the reproductive cycle, and evaluation of the effect of melatonin on hypothalamic and pituitary activities during the follicular and luteal phases of the estrous cycle. AANAT and melatonin receptors were localized in the pineal gland and preoptic area of the hypothalamus. Increase in pineal AANAT and serum melatonin expression was observed as pregnancy progressed, with the lowest hypothalamic MT1 and MT2 levels at mid-pregnancy. Pulsatility assays demonstrated that melatonin induces GnRH and LH secretion at luteal phase. The melatoninergic system effects on hypothalamic and pituitary gland hormones secretion during pregnancy pinpoint to melatonin as a potential key factor underlying the reactivation of the reproductive axis activity at mid-gestation.


Asunto(s)
Melatonina , Animales , Femenino , Hipotálamo/metabolismo , Hormona Luteinizante/metabolismo , Melatonina/metabolismo , Hipófisis/metabolismo , Embarazo , América del Sur
16.
Reprod Sci ; 28(12): 3547-3561, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33856666

RESUMEN

To explore in mice if a 15% food restriction protocol during pregnancy programs the offspring postnatal development, with emphasis on reproductive function, and to assess if ghrelin (Ghrl) administration to mouse dams exerts effects that mimic those obtained under mild caloric restriction. Mice were 15% food-restricted, injected with 4 nmol/animal/day of Ghrl, or injected with the vehicle (control) thorough pregnancy. After birth, the pups did not receive further treatment. Pups born from food-restricted dams (FR pups) were lighter than Ghrl pups at birth, but reached normal weight at adulthood. Ghrl pups were heavier at birth and gained more weight than control pups (C pups). This effect was not associated with plasma IGF-1. FR pups showed a delay in pinna detachment and eye opening, while an advance was observed in Ghrl pups. FR pups showed also impairment in the surface-righting reflex. In both female FR and Ghrl pups, there was an advance in vaginal opening and, in adulthood, FR pups showed a significant decrease in their own litter size and plasma progesterone, and an increase in embryo loss. A delay in testicular descent was evident in male Ghrl pups. Changes in puberty onset were not associated with differences in the expression of Kiss1 in hypothalamic nuclei. Finally, in adulthood, FR pups showed a significant decrease in sperm quality. In conclusion, a mild food restriction thorough gestation exerted programming effects on the offspring, affecting also their reproductive function in adulthood. These effects were not similar to those of intragestational Ghrl administration.


Asunto(s)
Restricción Calórica/métodos , Desarrollo Fetal/fisiología , Ghrelina/administración & dosificación , Efectos Tardíos de la Exposición Prenatal/genética , Desarrollo Sexual/fisiología , Animales , Animales Recién Nacidos , Vías de Administración de Medicamentos , Femenino , Desarrollo Fetal/efectos de los fármacos , Masculino , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Desarrollo Sexual/efectos de los fármacos
17.
Nucleic Acid Ther ; 31(2): 155-171, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33347786

RESUMEN

Type 1 diabetes occurs as a consequence of progressive autoimmune destruction of beta cells. A potential treatment for this disease should address the immune attack on beta cells and their preservation/regeneration. The objective of this study was to elucidate whether the immunomodulatory synthetic oligonucleotide IMT504 was able to ameliorate diabetes in NOD mice and to provide further understanding of its mechanism of action. We found that IMT504 restores glucose homeostasis in a diabetes mouse model similar to human type 1 diabetes, by regulating expression of immune modulatory factors and improving beta cell function. IMT504 treatment markedly improved fasting glycemia, insulinemia, and homeostatic model assessment of beta cell function (HOMA-Beta cell) index. Moreover, this treatment increased islet number and decreased apoptosis, insulitis, and CD45+ pancreas-infiltrating leukocytes. In a long-term treatment, we observed improvement of glucose metabolism up to 9 days after IMT504 cessation and increased survival after 15 days of the last IMT504 injection. We postulate that interleukin (IL)-12B (p40), possibly acting as a homodimer, and Galectin-3 (Gal-3) may function as mediators of this immunomodulatory action. Overall, these results validate the therapeutic activity of IMT504 as a promising drug for type 1 diabetes and suggest possible downstream mediators of its immunomodulatory effect.


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Insulina/genética , Oligodesoxirribonucleótidos/farmacología , Oligonucleótidos/farmacología , Animales , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Modelos Animales de Enfermedad , Femenino , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Ratones , Ratones Endogámicos NOD , Oligodesoxirribonucleótidos/genética , Oligonucleótidos/genética , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Páncreas/patología
18.
Am J Physiol Endocrinol Metab ; 298(3): E683-96, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20009027

RESUMEN

GABA, the main inhibitory neurotransmitter, acts through GABA(A/C) and GABA(B) receptors (GABA(B)Rs); it is critical for gonadotropin regulation. We studied whether the lack of functional GABA(B)Rs in GABA(B1) knockout (GABA(B1)KO) mice affected the gonadotropin axis physiology. Adult male and female GABA(B1)KO and wild-type (WT) mice were killed to collect blood and tissue samples. Gonadotropin-releasing hormone (GnRH) content in whole hypothalami (HT), olfactory bulbs (OB), and frontoparietal cortexes (CT) were determined (RIA). GnRH expression by quantitative real-time PCR (qRT-PCR) was evaluated in preoptic area-anterior hypothalamus (POA-AH), medial basal-posterior hypothalamus (MBH-PH), OB, and CT. Pulsatile GnRH secretion from hypothalamic explants was measured by RIA. GABA, glutamate, and taurine contents in HT and CT were determined by HPLC. Glutamic acid decarboxylase-67 (GAD-67) mRNA was measured by qRT-PCR in POA-AH, MBH-PH, and CT. Gonadotropin content, serum levels, and secretion from adenohypophyseal cell cultures (ACC) were measured by RIA. GnRH mRNA expression was increased in POA-AH of WT males compared with females; this pattern of expression was inversed in GABA(B1)KO mice. MBH-PH, OB, and CT did not follow this pattern. In GABA(B1)KO females, GnRH pulse frequency was increased and GABA and glutamate contents were augmented. POA-AH GAD-67 mRNA showed the same expression pattern as GnRH mRNA in this area. Gonadotropin pituitary contents and serum levels showed no differences between genotypes. Increased basal LH secretion and decreased GnRH-stimulated gonadotropin response were observed in GABA(B1)KO female ACCs. These results support the hypothesis that the absence of functional GABA(B)Rs alters GnRH physiology and critically affects sexual dimorphic expression of GnRH and GAD-67 in POA-AH.


Asunto(s)
Encéfalo/metabolismo , Glutamato Descarboxilasa/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Receptores de GABA-B/metabolismo , Caracteres Sexuales , Animales , Femenino , Expresión Génica/fisiología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Transducción de Señal/fisiología , Distribución Tisular
19.
J Steroid Biochem Mol Biol ; 200: 105627, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32070756

RESUMEN

The South American plains vizcacha, Lagostomus maximus, is the only mammal described so far that shows expression of estrogen receptors (ERs) and progesterone receptors (PRs) in gonadotropin-releasing hormone (GnRH) neurons. This animal therefore constitutes an exceptional model for the study of the effect of steroid hormones on the modulation of the hypothalamic-pituitary-ovarian (HPO) axis. By using both in vivo and ex vivo approaches, we have found that pharmacological doses of progesterone (P4) and estradiol (E2) produced an inhibition in the expression of hypothalamic GnRH, while physiological doses produced a differential effect on the pulsatile release frequency or genomic expression of GnRH. Our ex vivo experiment indicates that a short-term effect of E2 modulates the frequency of GnRH release pattern that would be associated with membrane ERs. On the other hand, our in vivo approach suggests that a long-term effect of E2, acting through the classical nuclear ERs-PRs pathway, would produce the modification of GnRH mRNA expression during the GnRH pre-ovulatory surge. Particularly, P4 induced a rise in GnRH mRNA expression and protein release with a decrease in its release frequency. These results suggest different levels of action of steroid hormones on GnRH modulation. We conclude that the fine action of E2 and P4 constitute the key factor to enable the hypothalamic activity during the pregnancy of this mammal.


Asunto(s)
Estradiol/farmacología , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/efectos de los fármacos , Progesterona/farmacología , Animales , Estradiol/sangre , Femenino , Hormona Liberadora de Gonadotropina/genética , Sistema Hipotálamo-Hipofisario , Hipotálamo/metabolismo , Hormona Luteinizante/metabolismo , Ovariectomía , Ovario , Progesterona/sangre , Roedores
20.
Brain Behav Immun ; 23(4): 464-73, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19486656

RESUMEN

Administration of psychostimulants can elicit a sensitized response to the stimulating and reinforcing properties of the drugs, although there is scarce information regarding their effects at immune level. We previously demonstrated that an acute exposure to amphetamine (5 mg/kg, i.p.) induced an inhibitory effect on the splenic T-cell proliferative response, along with an increase in met-enkephalin at limbic and immune levels, 4 days following drug administration. In this study, we evaluated the amphetamine-induced effects at weeks one and three after the same single dose treatment (5 mg/kg, i.p.) on the lymphoproliferative response and on the met-enkephalin in the nucleus accumbens (NAc), prefrontal cortex (PfC), spleen and thymus. It was demonstrated that these effects disappeared completely after three weeks, although re-exposure to an amphetamine challenge induced the expression of sensitization to the effects of amphetamine on the lymphoproliferative response and on the met-enkephalin from NAc, spleen and thymus, but not in the PfC. Pre-treatment with MK-801 (0.1 mg/kg, i.p.), an N-methyl-d-aspartate (NMDA) glutamatergic receptor antagonist, blocked the effects of a single amphetamine exposure on the lymphoproliferative response and on met-enkephalin in the NAc and spleen. Furthermore, the NMDA receptor antagonist administered prior to amphetamine challenge also blocked the expression of sensitization in both parameters evaluated. These findings show a long-lasting amphetamine-induced sensitization phenomenon at the immune level in a parallel way to that occurring in the limbic and immune enkephalineric system. A glutamate mechanism is implied in the long-term amphetamine-induced effects at immune level and in the met-enkephalin from NAc and spleen.


Asunto(s)
Anfetamina/administración & dosificación , Encefalina Metionina/metabolismo , Núcleo Accumbens/inmunología , Receptores de N-Metil-D-Aspartato/metabolismo , Bazo/inmunología , Análisis de Varianza , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Estimulantes del Sistema Nervioso Central/administración & dosificación , Maleato de Dizocilpina/administración & dosificación , Esquema de Medicación , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Ácido Glutámico/metabolismo , Masculino , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Radioinmunoensayo , Ratas , Ratas Wistar , Bazo/efectos de los fármacos , Bazo/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda