Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Am J Physiol Endocrinol Metab ; 315(3): E357-E366, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29812984

RESUMEN

Intraperitoneal administration of the melanocortin agonist melanotan II (MTII) to mice causes a profound, transient hypometabolism/hypothermia. It is preserved in mice lacking any one of melanocortin receptors 1, 3, 4, or 5, suggesting a mechanism independent of the canonical melanocortin receptors. Here we show that MTII-induced hypothermia was abolished in KitW-sh/W-sh mice, which lack mast cells, demonstrating that mast cells are required. MRGPRB2 is a receptor that detects many cationic molecules and activates mast cells in an antigen-independent manner. In vitro, MTII stimulated mast cells by both MRGPRB2-dependent and -independent mechanisms, and MTII-induced hypothermia was intact in MRGPRB2-null mice. Confirming that MTII activated mast cells, MTII treatment increased plasma histamine levels in both wild-type and MRGPRB2-null, but not in KitW-sh/W-sh, mice. The released histamine produced hypothermia via histamine H1 receptors because either a selective antagonist, pyrilamine, or ablation of H1 receptors greatly diminished the hypothermia. Other drugs, including compound 48/80, a commonly used mast cell activator, also produced hypothermia by both mast cell-dependent and -independent mechanisms. These results suggest that mast cell activation should be considered when investigating the mechanism of drug-induced hypothermia in mice.


Asunto(s)
Agonistas de los Receptores Histamínicos/farmacología , Hipotermia/inducido químicamente , Mastocitos/efectos de los fármacos , Péptidos Cíclicos/farmacología , alfa-MSH/análogos & derivados , Animales , Liberación de Histamina/efectos de los fármacos , Liberación de Histamina/genética , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , alfa-MSH/farmacología
2.
J Pharmacol Exp Ther ; 356(2): 474-82, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26606937

RESUMEN

Adenosine can induce hypothermia, as previously demonstrated for adenosine A1 receptor (A1AR) agonists. Here we use the potent, specific A3AR agonists MRS5698, MRS5841, and MRS5980 to show that adenosine also induces hypothermia via the A3AR. The hypothermic effect of A3AR agonists is independent of A1AR activation, as the effect was fully intact in mice lacking A1AR but abolished in mice lacking A3AR. A3AR agonist-induced hypothermia was attenuated by mast cell granule depletion, demonstrating that the A3AR hypothermia is mediated, at least in part, via mast cells. Central agonist dosing had no clear hypothermic effect, whereas peripheral dosing of a non-brain-penetrant agonist caused hypothermia, suggesting that peripheral A3AR-expressing cells drive the hypothermia. Mast cells release histamine, and blocking central histamine H1 (but not H2 or H4) receptors prevented the hypothermia. The hypothermia was preceded by hypometabolism and mice with hypothermia preferred a cooler environmental temperature, demonstrating that the hypothermic state is a coordinated physiologic response with a reduced body temperature set point. Importantly, hypothermia is not required for the analgesic effects of A3AR agonists, which occur with lower agonist doses. These results support a mechanistic model for hypothermia in which A3AR agonists act on peripheral mast cells, causing histamine release, which stimulates central histamine H1 receptors to induce hypothermia. This mechanism suggests that A3AR agonists will probably not be useful for clinical induction of hypothermia.


Asunto(s)
Hipotermia/metabolismo , Receptor de Adenosina A3/metabolismo , Receptores Histamínicos H1/metabolismo , Agonistas del Receptor de Adenosina A3/farmacología , Animales , Relación Dosis-Respuesta a Droga , Hipotermia/inducido químicamente , Masculino , Mastocitos/efectos de los fármacos , Mastocitos/metabolismo , Ratones , Ratones Endogámicos C57BL
3.
J Neurosci ; 34(18): 6182-9, 2014 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-24790189

RESUMEN

Locus ceruleus (LC) noradrenergic neurons are critical in generating alertness. In addition to inducing cortical arousal, the LC also orchestrates changes in accompanying autonomic system function that compliments increased attention, such as during stress, excitation, and/or exposure to averse or novel stimuli. Although the association between arousal and increased heart rate is well accepted, the neurobiological link between the LC and parasympathetic neurons that control heart rate has not been identified. In this study, we test directly whether activation of noradrenergic neurons in the LC influences brainstem parasympathetic cardiac vagal neurons (CVNs). CVNs were identified in transgenic mice that express channel-rhodopsin-2 (ChR2) in LC tyrosine hydroxylase neurons. Photoactivation evoked a rapid depolarization, increased firing, and excitatory inward currents in ChR2-expressing neurons in the LC. Photostimulation of LC neurons did not alter excitatory currents, but increased inhibitory neurotransmission to CVNs. Optogenetic activation of LC neurons increased the frequency of isolated glycinergic IPSCs by 27 ± 8% (p = 0.003, n = 26) and augmented GABAergic IPSCs in CVNs by 21 ± 5% (p = 0.001, n = 26). Inhibiting α1, but not α2, receptors blocked the evoked responses. Inhibiting ß1 receptors prevented the increase in glycinergic, but not GABAergic, IPSCs in CVNs. This study demonstrates LC noradrenergic neurons inhibit the brainstem CVNs that generate parasympathetic activity to the heart. This inhibition of CVNs would increase heart rate and risks associated with tachycardia. The receptors activated within this pathway, α1 and/or ß1 receptors, are targets for clinically prescribed antagonists that promote slower, cardioprotective heart rates during heightened vigilant states.


Asunto(s)
Tronco Encefálico/citología , Locus Coeruleus/citología , Neuronas/fisiología , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Nervio Vago/fisiología , Adrenérgicos/farmacología , Vías Aferentes/fisiología , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Channelrhodopsins , Glicina/farmacología , Corazón/inervación , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/fisiología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Optogenética , Técnicas de Placa-Clamp , Estimulación Luminosa , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
4.
Cell Metab ; 34(2): 285-298.e7, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35108515

RESUMEN

The central nervous system has long been thought to regulate insulin secretion, an essential process in the maintenance of blood glucose levels. However, the anatomical and functional connections between the brain and insulin-producing pancreatic ß cells remain undefined. Here, we describe a functional transneuronal circuit connecting the hypothalamus to ß cells in mice. This circuit originates from a subpopulation of oxytocin neurons in the paraventricular hypothalamic nucleus (PVNOXT), and it reaches the islets of the endocrine pancreas via the sympathetic autonomic branch to innervate ß cells. Stimulation of PVNOXT neurons rapidly suppresses insulin secretion and causes hyperglycemia. Conversely, silencing of these neurons elevates insulin levels by dysregulating neuronal signaling and secretory pathways in ß cells and induces hypoglycemia. PVNOXT neuronal activity is triggered by glucoprivation. Our findings reveal that a subset of PVNOXT neurons form functional multisynaptic circuits with ß cells in mice to regulate insulin secretion, and their function is necessary for the ß cell response to hypoglycemia.


Asunto(s)
Células Secretoras de Insulina , Animales , Hipotálamo/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Ratones , Oxitocina/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo
5.
eNeuro ; 8(4)2021.
Artículo en Inglés | MEDLINE | ID: mdl-34326065

RESUMEN

Bombesin receptor subtype-3 (BRS3) is an orphan receptor that regulates energy homeostasis. We compared Brs3 driver mice with constitutive or inducible Cre recombinase activity. The constitutive BRS3-Cre mice show a reporter signal (Cre-dependent tdTomato) in the adult brain because of lineage tracing in the dentate gyrus, striatal patches, and indusium griseum, in addition to sites previously identified in the inducible BRS3-Cre mice (including hypothalamic and amygdala subregions, and parabrachial nucleus). We detected Brs3 reporter expression in the dentate gyrus at day 23 but not at postnatal day 1 or 5 months of age. Hypothalamic sites expressed reporter at all three time points, and striatal patches expressed Brs3 reporter at 1 day but not 5 months. Parabrachial nucleus Brs3 neurons project to the preoptic area, hypothalamus, amygdala, and thalamus. Both Cre recombinase insertions reduced Brs3 mRNA levels and BRS3 function, causing obesity phenotypes of different severity. These results demonstrate that driver mice should be characterized phenotypically and illustrate the need for knock-in strategies with less effect on the endogenous gene.


Asunto(s)
Integrasas , Receptores de Bombesina , Animales , Encéfalo/metabolismo , Hipotálamo/metabolismo , Integrasas/genética , Integrasas/metabolismo , Ratones , Ratones Transgénicos , Receptores de Bombesina/metabolismo
6.
Cell Metab ; 33(7): 1389-1403.e6, 2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34038711

RESUMEN

The preoptic area (POA) is a key brain region for regulation of body temperature (Tb), dictating thermogenic, cardiovascular, and behavioral responses that control Tb. Previously characterized POA neuronal populations all reduced Tb when activated. Using mice, we now identify POA neurons expressing bombesin-like receptor 3 (POABRS3) as a population whose activation increased Tb; inversely, acute inhibition of these neurons reduced Tb. POABRS3 neurons that project to either the paraventricular nucleus of the hypothalamus or the dorsomedial hypothalamus increased Tb, heart rate, and blood pressure via the sympathetic nervous system. Long-term inactivation of POABRS3 neurons caused increased Tb variability, overshooting both increases and decreases in Tb set point, with RNA expression profiles suggesting multiple types of POABRS3 neurons. Thus, POABRS3 neuronal populations regulate Tb and heart rate, contribute to cold defense, and fine-tune feedback control of Tb. These findings advance understanding of homeothermy, a defining feature of mammalian biology.


Asunto(s)
Regulación de la Temperatura Corporal , Frecuencia Cardíaca , Neuronas/fisiología , Área Preóptica/metabolismo , Receptores de Bombesina/metabolismo , Animales , Temperatura Corporal/genética , Regulación de la Temperatura Corporal/genética , Frecuencia Cardíaca/genética , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/citología , Neuronas/metabolismo , Área Preóptica/citología , Receptores de Bombesina/genética , Transducción de Señal/genética , Sistema Nervioso Simpático/fisiología , Termogénesis/genética
7.
Mol Metab ; 36: 100969, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32229422

RESUMEN

OBJECTIVE: Bombesin-like receptor 3 (BRS3) is an orphan receptor and Brs3 knockout mice develop obesity with increased food intake and reduced resting metabolic rate and body temperature. The neuronal populations contributing to these effects were examined. METHODS: We studied energy metabolism in mice with Cre-mediated recombination causing 1) loss of BRS3 selectively in SIM1- or MC4R-expressing neurons or 2) selective re-expression of BRS3 from a null background in these neurons. RESULTS: The deletion of BRS3 in MC4R neurons increased body weight/adiposity, metabolic efficiency, and food intake, and reduced insulin sensitivity. BRS3 re-expression in these neurons caused partial or no reversal of these traits. However, these observations were confounded by an obesity phenotype caused by the Mc4r-Cre allele, independent of its recombinase activity. The deletion of BRS3 in SIM1 neurons increased body weight/adiposity and food intake, but not to the levels of the global null. The re-expression of BRS3 in SIM1 neurons reduced body weight/adiposity and food intake, but not to wild type levels. The deletion of BRS3 in either MC4R- or SIM1-expressing neurons affected body temperature, with re-expression in either population reversing the null phenotype. MK-5046, a BRS3 agonist, increases light phase body temperature in wild type, but not Brs3 null, mice and BRS3 re-expression in either population restored response to MK-5046. CONCLUSIONS: BRS3 in both MC4R- and SIM1-expressing neurons contributes to regulation of body weight/adiposity, insulin sensitivity, food intake, and body temperature.


Asunto(s)
Metabolismo Energético/fisiología , Neuronas/metabolismo , Receptores de Bombesina/metabolismo , Adiposidad/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Temperatura Corporal/fisiología , Peso Corporal , Encéfalo/metabolismo , Ingestión de Alimentos/fisiología , Femenino , Homeostasis/fisiología , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/fisiología , Obesidad/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , Receptores de Bombesina/genética , Proteínas Represoras/metabolismo
8.
PLoS One ; 15(12): e0243986, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33326493

RESUMEN

Extracellular adenosine, a danger signal, can cause hypothermia. We generated mice lacking neuronal adenosine A1 receptors (A1AR, encoded by the Adora1 gene) to examine the contribution of these receptors to hypothermia. Intracerebroventricular injection of the selective A1AR agonist (Cl-ENBA, 5'-chloro-5'-deoxy-N6-endo-norbornyladenosine) produced hypothermia, which was reduced in mice with deletion of A1AR in neurons. A non-brain penetrant A1AR agonist [SPA, N6-(p-sulfophenyl) adenosine] also caused hypothermia, in wild type but not mice lacking neuronal A1AR, suggesting that peripheral neuronal A1AR can also cause hypothermia. Mice expressing Cre recombinase from the Adora1 locus were generated to investigate the role of specific cell populations in body temperature regulation. Chemogenetic activation of Adora1-Cre-expressing cells in the preoptic area did not change body temperature. In contrast, activation of Adora1-Cre-expressing dorsomedial hypothalamus cells increased core body temperature, concordant with agonism at the endogenous inhibitory A1AR causing hypothermia. These results suggest that A1AR agonism causes hypothermia via two distinct mechanisms: brain neuronal A1AR and A1AR on neurons outside the blood-brain barrier. The variety of mechanisms that adenosine can use to induce hypothermia underscores the importance of hypothermia in the mouse response to major metabolic stress or injury.


Asunto(s)
Hipotermia/metabolismo , Receptor de Adenosina A1/metabolismo , Agonistas del Receptor de Adenosina A1/farmacología , Animales , Hipotálamo/metabolismo , Hipotálamo/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Nervios Periféricos/metabolismo , Nervios Periféricos/fisiopatología
9.
Brain Res ; 1201: 88-92, 2008 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-18295749

RESUMEN

Recent work has shown that adenosine 5'-triphosphate (ATP) plays an important role in modulating the activity of parasympathetic cardiac vagal neurons that dominate the neural control of heart rate. This study examined the mechanisms by which activation of ATP receptors modulates excitatory neurotransmission to cardiac vagal neurons. Glutamatergic activity to cardiac vagal neurons was isolated and examined using whole-cell patch-clamp recordings in an in vitro brain slice preparation in rats. ATP (100 microM) evoked increases in the frequency of glutamatergic miniature excitatory postsynaptic currents (mEPSCs) in cardiac vagal neurons which were blocked by the broad P2 receptor antagonist pyridoxal-phosphate-6-azophenyl-2',4'-disulphonic acid (PPADS, 100 microM). Application of the selective P2X receptor agonist, alpha, beta-methylene ATP (100 microM), also increased glutamatergic mEPSCs neurotransmission to cardiac vagal neurons indicating P2X receptors enhance glutamatergic release to cardiac vagal neurons. The evoked increase in glutamatergic mEPSC was unaltered by the voltage-gated calcium channel blocker cadmium, and was abolished by the selective P2X receptor antagonist 2',3'-O-(2,4,6-Trinitrophenyl) adenosine 5'-triphosphate, TNP-ATP (100 microM). This work demonstrates that the ATP evoked facilitation of excitatory neurotransmission to cardiac vagal neurons is dependent upon activation of P2X receptors on glutamatergic presynaptic terminals.


Asunto(s)
Adenosina Trifosfato/fisiología , Ácido Glutámico/metabolismo , Bulbo Raquídeo/metabolismo , Neuronas/metabolismo , Transmisión Sináptica/fisiología , Nervio Vago/metabolismo , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Animales , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Ganglios Parasimpáticos/fisiología , Corazón/inervación , Bulbo Raquídeo/efectos de los fármacos , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Sistema Nervioso Parasimpático/efectos de los fármacos , Sistema Nervioso Parasimpático/metabolismo , Técnicas de Placa-Clamp , Agonistas del Receptor Purinérgico P2 , Antagonistas del Receptor Purinérgico P2 , Fosfato de Piridoxal/análogos & derivados , Fosfato de Piridoxal/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato/efectos de los fármacos , Receptores de Glutamato/metabolismo , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2X , Transmisión Sináptica/efectos de los fármacos , Nervio Vago/efectos de los fármacos
10.
Brain Res ; 1224: 53-62, 2008 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-18590708

RESUMEN

This study examined whether adenosine 5'-triphosphate (ATP) modulated inhibitory glycinergic and GABAergic neurotransmission to cardiac vagal neurons. Inhibitory activity to cardiac vagal neurons was isolated and examined using whole-cell patch-clamp recordings in an in vitro brain slice preparation in rats. ATP (100 microM) evoked increases in the frequency of glycinergic and GABAergic miniature inhibitory postsynaptic currents (mIPSCs) in cardiac vagal neurons which were blocked by the broad P2 receptor antagonist pyridoxal-phosphate-6-azophenyl-2',4'-disulphonic acid (100 microM). Application of the P2Y agonists uridine triphosphate (15 microM) and adenosine 5'-0-(Z-thiodiphosphate) (60 microM) did not enhance inhibitory neurotransmission to cardiac vagal neurons however, application of the selective P2X; receptor agonist, alpha, beta-methylene ATP (100 microM), increased glycinergic and GABAergic mIPSC neurotransmission to cardiac vagal neurons. The increase in inhibitory neurotransmission evoked by alpha, beta-methylene ATP was abolished by the selective P2X receptor antagonist 2',3'-O-(2,4,6-Trinitrophenyl) adenosine 5'-triphosphate (100 microM) indicating P2X receptors enhance the release of inhibitory neurotransmitters to cardiac neurons. The voltage-gated calcium channel blocker cadmium chloride did not alter the evoked increase in inhibitory mIPSCs. This work demonstrates that P2X receptor activation enhances inhibitory neurotransmission to parasympathetic cardiac vagal neurons and demonstrates an important functional role for ATP mediated purinergic signaling to cardiac vagal neurons.


Asunto(s)
Corazón/inervación , Bulbo Raquídeo/metabolismo , Inhibición Neural/fisiología , Neuronas/metabolismo , Receptores Purinérgicos P2/metabolismo , Nervio Vago/metabolismo , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacología , Animales , Animales Recién Nacidos , Bloqueadores de los Canales de Calcio/farmacología , Glicina/metabolismo , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/fisiología , Bulbo Raquídeo/citología , Inhibición Neural/efectos de los fármacos , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Agonistas del Receptor Purinérgico P2 , Antagonistas del Receptor Purinérgico P2 , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos P2X , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Nervio Vago/citología , Ácido gamma-Aminobutírico/metabolismo
11.
Nat Neurosci ; 21(11): 1530-1540, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30349101

RESUMEN

Bombesin-like receptor 3 (BRS3) is an orphan G-protein-coupled receptor that regulates energy homeostasis and heart rate. We report that acute activation of Brs3-expressing neurons in the dorsomedial hypothalamus (DMHBrs3) increased body temperature (Tb), brown adipose tissue temperature, energy expenditure, heart rate, and blood pressure, with no effect on food intake or physical activity. Conversely, activation of Brs3 neurons in the paraventricular nucleus of the hypothalamus had no effect on Tb or energy expenditure, but suppressed food intake. Inhibition of DMHBrs3 neurons decreased Tb and energy expenditure, suggesting a necessary role in Tb regulation. We found that the preoptic area provides major input (excitatory and inhibitory) to DMHBrs3 neurons. Optogenetic stimulation of DMHBrs3 projections to the raphe pallidus increased Tb. Thus, DMHBrs3→raphe pallidus neurons regulate Tb, energy expenditure, and heart rate, and Brs3 neurons in the paraventricular nucleus of the hypothalamus regulate food intake. Brs3 expression is a useful marker for delineating energy metabolism regulatory circuitry.


Asunto(s)
Temperatura Corporal/fisiología , Núcleo Hipotalámico Dorsomedial/metabolismo , Ingestión de Alimentos/fisiología , Metabolismo Energético/fisiología , Frecuencia Cardíaca/fisiología , Neuronas/metabolismo , Receptores de Bombesina/metabolismo , Animales , Masculino , Ratones
12.
Temperature (Austin) ; 9(4): 306-309, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36339094
13.
Mol Metab ; 6(11): 1540-1550, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29107299

RESUMEN

OBJECTIVE: Bombesin-like receptor 3 (BRS-3) is an orphan G protein-coupled receptor. Brs3 null mice have reduced resting metabolic rate and body temperature, increased food intake, and obesity. Here we study the role of Brs3 in different neuron types. METHODS: Mice able to undergo Cre recombinase-dependent inactivation or re-expression of Brs3 were generated, respectively Brs3fl/y and Brs3loxTB/y. We then studied four groups of mice with Brs3 selectively inactivated or re-expressed in cells expressing Vglut2-Cre or Vgat-Cre. RESULTS: Deletion of Brs3 in glutamatergic neurons expressing Vglut2 reproduced the global null phenotype for regulation of food intake, metabolic rate, body temperature, adiposity, and insulin resistance. These mice also no longer responded to a BRS-3 agonist, MK-5046. In contrast, deletion of Brs3 in GABAergic neurons produced no detectable phenotype. Conversely, the wild type phenotype was restored by selective re-expression of Brs3 in glutamatergic neurons, with no normalization achieved by re-expressing Brs3 in GABAergic neurons. CONCLUSIONS: Brs3 expression in glutamatergic neurons is both necessary and sufficient for full Brs3 function in energy metabolism. In these experiments, no function was identified for Brs3 in GABAergic neurons. The data suggest that the anti-obesity pharmacologic actions of BRS-3 agonists occur via agonism of receptors on glutamatergic neurons.


Asunto(s)
Ácido Glutámico/metabolismo , Neuronas/metabolismo , Receptores de Bombesina/biosíntesis , Adiposidad , Animales , Peso Corporal , Ingestión de Alimentos/fisiología , Metabolismo Energético , Neuronas GABAérgicas/metabolismo , Expresión Génica , Imidazoles/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Obesidad/metabolismo , Pirazoles/farmacología , Receptores de Bombesina/agonistas , Receptores de Bombesina/genética
14.
Cell Res ; 29(10): 785-786, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31471559
15.
Hypertension ; 64(3): 597-603, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24958501

RESUMEN

Obstructive sleep apnea is associated with chronic intermittent hypoxia/hypercapnia (CIHH) episodes during sleep that heighten sympathetic and diminish parasympathetic activity to the heart. Although one population of neurons in the paraventricular nucleus of the hypothalamus strongly influences sympathetic tone and has increased activity after CIHH, little is known about the role of this pathway to parasympathetic neurons and how this network is altered in CIHH. We hypothesized that CIHH inhibits the excitatory pathway from the paraventricular nucleus of the hypothalamus to parasympathetic cardiac vagal neurons in the brain stem. To test this hypothesis, channelrhodopsin was selectively expressed, using viral vectors, in neurons in the paraventricular nucleus of the hypothalamus and channelrhodopsin-expressing fibers were photoactivated to evoke postsynaptic currents in cardiac vagal neurons in brain stem slices. Excitatory postsynaptic currents were diminished in animals exposed to CIHH. The paired-pulse and prolonged facilitation of the postsynaptic current amplitudes and frequencies evoked by paired and bursts of photoactivation of channelrhodopsin fibers, respectively, occurred in unexposed rats but were blunted in CIHH animals. In response to an acute challenge of hypoxia/hypercapnia, the amplitude of postsynaptic events was unchanged during, but increased after hypoxia/hypercapnia in unexposed animals. In contrast, postsynaptic currents were inhibited during hypoxia/hypercapnia in rats exposed to CIHH. In conclusion, the excitatory pathway to cardiac vagal neurons is diminished in response to both acute and chronic exposures to hypoxia/hypercapnia. This could elicit a reduced cardioprotective parasympathetic activity and an enhanced risk of adverse cardiovascular events in episodes of apnea and chronic obstructive sleep apnea.


Asunto(s)
Tronco Encefálico/fisiopatología , Corazón/inervación , Hipercapnia/fisiopatología , Hipoxia/fisiopatología , Sistema Nervioso Parasimpático/fisiopatología , Núcleo Hipotalámico Paraventricular/fisiopatología , Animales , Enfermedades Cardiovasculares/epidemiología , Modelos Animales de Enfermedad , Femenino , Masculino , Proteínas del Tejido Nervioso/fisiología , Ratas , Ratas Sprague-Dawley , Factores de Riesgo , Apnea Obstructiva del Sueño/fisiopatología , Transmisión Sináptica/fisiología
16.
PLoS One ; 9(11): e112138, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25379676

RESUMEN

Recent work has shown that oxytocin is involved in more than lactation and uterine contraction. The paraventricular nucleus of the hypothalamus (PVN) contains neuroendocrine neurons that control the release of hormones, including vasopressin and oxytocin. Other populations of PVN neurons do not release hormones, but rather project to and release neurotransmitters onto other neurons in the CNS involved in fluid retention, thermoregulation, sexual behavior and responses to stress. Activation of oxytocin receptors can be cardioprotective and reduces the adverse cardiovascular consequences of anxiety and stress, yet how oxytocin can affect heart rate and cardiac function is unknown. While anatomical work has shown the presence of peptides, including oxytocin, in the projections from the PVN to parasympathetic nuclei, electrophysiological studies to date have only demonstrated release of glutamate and activation of fast ligand gated receptors in these pathways. In this study, using rats, we directly show, using sniffer CHO cells that express oxytocin receptors and the Ca2+ indicator R-GECO, that optogenetic activation of channelrhodopsin-2 (ChR2) expressing PVN fibers in the brainstem activates oxytocin receptors in the dorsomotor nucleus of the vagus (DMNV). We also demonstrate that while a single photoactivation of PVN terminals only activates glutamatergic receptors in brainstem cardiac vagal neurons (CVNs), neurons that dominate the neural control of heart rate, both the paired pulse facilitation, and sustained enhancement of glutamate release in this pathway is mediated by activation of oxytocin receptors. Our results provide direct evidence that a pathway from the PVN likely releases oxytocin and enhances short-term plasticity of this critical autonomic connection.


Asunto(s)
Tronco Encefálico/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Oxitocina/metabolismo , Animales , Tronco Encefálico/citología , Células CHO , Cricetulus , Femenino , Hipotálamo/citología , Masculino , Vías Nerviosas/citología , Vías Nerviosas/metabolismo , Neuronas/citología , Oxitocina/análisis , Ratas Sprague-Dawley , Receptores de Oxitocina/análisis , Receptores de Oxitocina/metabolismo
17.
Cell Metab ; 20(2): 333-45, 2014 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-24981835

RESUMEN

The melanocortin system regulates metabolic homeostasis and inflammation. Melanocortin agonists have contradictorily been reported to both increase and decrease metabolic rate and body temperature. We find two distinct physiologic responses occurring at similar doses. Intraperitoneal administration of the nonselective melanocortin agonist MTII causes a melanocortin-4 receptor (Mc4r)-mediated hypermetabolism/hyperthermia. This is preceded by a profound, transient hypometabolism/hypothermia that is preserved in mice lacking any one of Mc1r, Mc3r, Mc4r, or Mc5r. Three other melanocortin agonists also caused hypothermia, which is actively achieved via seeking a cool environment, vasodilation, and inhibition of brown adipose tissue thermogenesis. These results suggest that the hypometabolic/hypothermic effect of MTII is not due to a failure of thermoregulation. The hypometabolism/hypothermia was prevented by dopamine antagonists, and MTII selectively activated arcuate nucleus dopaminergic neurons, suggesting that these neurons may contribute to the hypometabolism/hypothermia. We propose that the hypometabolism/hypothermia is a regulated response, potentially beneficial during extreme physiologic stress.


Asunto(s)
Temperatura Corporal/efectos de los fármacos , Receptores de Melanocortina/agonistas , alfa-MSH/análogos & derivados , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Antagonistas de Dopamina/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Metabolismo Energético/efectos de los fármacos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Melanocortina Tipo 1/agonistas , Receptor de Melanocortina Tipo 1/genética , Receptor de Melanocortina Tipo 1/metabolismo , Receptor de Melanocortina Tipo 3/agonistas , Receptor de Melanocortina Tipo 3/genética , Receptor de Melanocortina Tipo 3/metabolismo , Receptor de Melanocortina Tipo 4/agonistas , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , Receptores de Melanocortina/genética , Receptores de Melanocortina/metabolismo , alfa-MSH/farmacología
19.
J Neurosci Methods ; 210(2): 238-46, 2012 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-22890236

RESUMEN

Recent advances in optogenetic methods demonstrate the feasibility of selective photoactivation at the soma of neurons that express channelrhodopsin-2 (ChR2), but a comprehensive evaluation of different methods to selectively evoke transmitter release from distant synapses using optogenetic approaches is needed. Here we compared different lentiviral vectors, with sub-population-specific and strong promoters, and transgenic methods to express and photostimulate ChR2 in the long-range projections of paraventricular nucleus of the hypothalamus (PVN) neurons to brain stem cardiac vagal neurons (CVNs). Using PVN subpopulation-specific promoters for vasopressin and oxytocin, we were able to depolarize the soma of these neurons upon photostimulation, but these promoters were not strong enough to drive sufficient expression for optogenetic stimulation and synaptic release from the distal axons. However, utilizing the synapsin promoter photostimulation of distal PVN axons successfully evoked glutamatergic excitatory post-synaptic currents in CVNs. Employing the Cre/loxP system, using the Sim-1 Cre-driver mouse line, we found that the Rosa-CAG-LSL-ChR2-EYFP Cre-responder mice expressed higher levels of ChR2 than the Rosa-CAG-LSL-ChR2-tdTomato line in the PVN, judged by photo-evoked currents at the soma. However, neither was able to drive sufficient expression to observe and photostimulate the long-range projections to brainstem autonomic regions. We conclude that a viral vector approach with a strong promoter is required for successful optogenetic stimulation of distal axons to evoke transmitter release in pre-autonomic PVN neurons. This approach can be very useful to study important hypothalamus-brainstem connections, and can be easily modified to selectively activate other long-range projections within the brain.


Asunto(s)
Tronco Encefálico/citología , Hipotálamo/citología , Vías Nerviosas/fisiología , Optogenética , Sinapsis/fisiología , 6-Ciano 7-nitroquinoxalina 2,3-diona/farmacología , Factores de Edad , Animales , Animales Recién Nacidos , Proteínas Bacterianas/genética , Biofisica , Channelrhodopsins , Estimulación Eléctrica , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Ganglios Autónomos/fisiología , Vectores Genéticos/genética , Corazón/inervación , Humanos , Técnicas In Vitro , Proteínas Luminiscentes/genética , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Oxitocina/genética , Oxitocina/metabolismo , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Bloqueadores de los Canales de Sodio/farmacología , Sinapsis/metabolismo , Sinapsinas/genética , Sinapsinas/metabolismo , Tetrodotoxina/farmacología , Transducción Genética/métodos , Valina/análogos & derivados , Valina/farmacología , Vasopresinas/genética , Vasopresinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA