Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Cell Neurosci ; 126: 103863, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37268282

RESUMO

Glucose accesses the brain primarily via the astrocyte cell compartment, where it passes through the glycogen shunt before catabolism to the oxidizable fuel L-lactate. Glycogen phosphorylase (GP) isoenzymes GPbb and GPmm impose distinctive control of ventromedial hypothalamic nucleus (VMN) glucose-regulatory neurotransmission during hypoglycemia, but lactate and/or gliotransmitter involvement in those actions is unknown. Lactate or the octadecaneuropeptide receptor antagonist cyclo(1-8)[DLeu5] OP (LV-1075) did not affect gene product down-regulation caused by GPbb or GPmm siRNA, but suppressed non-targeted GP variant expression in a VMN region-specific manner. Hypoglycemic up-regulation of neuronal nitric oxide synthase was enhanced in rostral and caudal VMN by GPbb knockdown, yet attenuated by GPMM siRNA in the middle VMN; lactate or LV-1075 reversed these silencing effects. Hypoglycemic inhibition of glutamate decarboxylase65/67 was magnified by GPbb (middle and caudal VMN) or GPmm (middle VMN) knockdown, responses that were negated by lactate or LV-1075. GPbb or GPmm siRNA enlarged hypoglycemic VMN glycogen profiles in rostral and middle VMN. Lactate and LV-1075 elicited progressive rostral VMN glycogen augmentation in GPbb knockdown rats, but stepwise-diminution of rostral and middle VMN glycogen after GPmm silencing. GPbb, not GPmm, knockdown caused lactate or LV-1075 - reversible amplification of hypoglycemic hyperglucagonemia and hypercorticosteronemia. Results show that lactate and octadecaneuropeptide exert opposing control of GPbb protein in distinct VMN regions, while the latter stimulates GPmm. During hypoglycemia, GPbb and GPmm may respectively diminish (rostral, caudal VMN) or enhance (middle VMN) nitrergic transmission and each oppose GABAergic signaling (middle VMN) by lactate- and octadecaneuropeptide-dependent mechanisms.


Assuntos
Hipoglicemia , Núcleo Hipotalâmico Ventromedial , Ratos , Animais , Núcleo Hipotalâmico Ventromedial/metabolismo , Isoenzimas/metabolismo , Ratos Sprague-Dawley , Hipoglicemia/metabolismo , Glucose/metabolismo , Glicogênio/metabolismo , Hipoglicemiantes/metabolismo , Hipoglicemiantes/farmacologia , Neurotransmissores/farmacologia , Glicogênio Fosforilase/metabolismo , Glicogênio Fosforilase/farmacologia , Lactatos/metabolismo , Lactatos/farmacologia , Hormônios/metabolismo , Hormônios/farmacologia
2.
Biology (Basel) ; 12(2)2023 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-36829519

RESUMO

The enzyme aromatase is expressed at high levels in the ventromedial hypothalamic nucleus (VMN), a principal component of the brain gluco-regulatory network. Current research utilized selective gene knockdown tools to investigate the premise that VMN neuroestradiol controls glucostasis. Intra-VMN aromatase siRNA administration decreased baseline aromatase protein expression and tissue estradiol concentrations and either reversed or attenuated the hypoglycemic regulation of these profiles in a VMN segment-specific manner. Aromatase gene repression down-regulated protein biomarkers for gluco-stimulatory (nitric oxide; NO) and -inhibitory (gamma-aminobutyric acid; GABA) neurochemical transmitters. Insulin-induced hypoglycemia (IIH) up- or down-regulated neuronal nitric oxide synthase (nNOS) and glutamate decarboxylase65/67 (GAD), respectively, throughout the VMN. Interestingly, IIH caused divergent changes in tissue aromatase and estradiol levels in rostral (diminished) versus middle and caudal (elevated) VMN. Aromatase knockdown prevented hypoglycemic nNOS augmentation in VMN middle and caudal segments, but abolished the GAD inhibitory response to IIH throughout this nucleus. VMN nitrergic and GABAergic neurons monitor stimulus-specific glycogen breakdown. Here, glycogen synthase (GS) and phosphorylase brain- (GPbb; AMP-sensitive) and muscle- (GPmm; noradrenergic -responsive) type isoform responses to aromatase siRNA were evaluated. Aromatase repression reduced GPbb and GPmm content in euglycemic controls and prevented hypoglycemic regulation of GPmm but not GPbb expression while reversing glycogen accumulation. Aromatase siRNA elevated baseline glucagon and corticosterone secretion and abolished hypoglycemic hyperglucagonemia and hypercorticosteronemia. Outcomes document the involvement of VMN neuroestradiol signaling in brain control of glucose homeostasis. Aromatase regulation of VMN gluco-regulatory signaling of hypoglycemia-associated energy imbalance may entail, in part, control of GP variant-mediated glycogen disassembly.

3.
ASN Neuro ; 13: 17590914211035020, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34596459

RESUMO

Brain glycogen is remodeled during metabolic homeostasis and provides oxidizable L-lactate equivalents. Brain glycogen phosphorylase (GP)-brain (GPbb; AMP-sensitive) and -muscle (GPmm; norepinephrine-sensitive) type isoforms facilitate stimulus-specific control of glycogen disassembly. Here, a whole animal model involving stereotactic-targeted delivery of GPmm or GPbb siRNA to the ventromedial hypothalamic nucleus (VMN) was used to investigate the premise that these variants impose differential control of gluco-regulatory transmission. Intra-VMN GPmm or GPbb siRNA administration inhibited glutamate decarboxylate65/67 (GAD), a protein marker for the gluco-inhibitory transmitter γ--aminobutyric acid (GABA), in the caudal VMN. GPbb knockdown, respectively overturned or exacerbated hypoglycemia-associated GAD suppression in rostral and caudal VMN. GPmm siRNA caused a segment-specific reversal of hypoglycemic augmentation of the gluco-stimulatory transmitter indicator, neuronal nitric oxide synthase (nNOS). In both cell types, GP siRNA down-regulated 5'-AMP-activated protein kinase (AMPK) during euglycemia, but hypoglycemic suppression of AMPK was reversed by GPmm targeting. GP knockdown elevated baseline GABA neuron phosphoAMPK (pAMKP) content, and amplified hypoglycemic augmentation of pAMPK expression in each neuron type. GPbb knockdown increased corticosterone secretion in eu- and hypoglycemic rats. Outcomes validate efficacy of GP siRNA delivery for manipulation of glycogen breakdown in discrete brain structures in vivo, and document VMN GPbb control of local GPmm expression. Results document GPmm and/or -bb regulation of GABAergic and nitrergic transmission in discrete rostro-caudal VMN segments. Contrary effects of glycogenolysis on metabolic-sensory AMPK protein during eu- versus hypoglycemia may reflect energy state-specific astrocyte signaling. Amplifying effects of GPbb knockdown on hypoglycemic stimulation of pAMPK infer that glycogen mobilization by GPbb limits neuronal energy instability during hypoglycemia.


Assuntos
Proteínas Quinases Ativadas por AMP , Núcleo Hipotalâmico Ventromedial , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Glicogênio Fosforilase/genética , Neurônios/metabolismo , Isoformas de Proteínas , Ratos , Ratos Sprague-Dawley , Núcleo Hipotalâmico Ventromedial/metabolismo
4.
Acta Neurobiol Exp (Wars) ; 81(2): 196-206, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34170267

RESUMO

Glycogen metabolism shapes ventromedial hypothalamic nucleus (VMN) control of glucose homeostasis. Brain glycogen mass undergoes compensatory expansion post­recovery from insulin­induced hypoglycemia (IIH). Current research utilized combinatory high­resolution microdissection/high­sensitivity Western blotting to investigate whether IIH causes residual adjustments in glycogen metabolism within the metabolic­sensory ventrolateral VMN (VMNvl). Micropunch­dissected tissue was collected from rostral, middle, and caudal levels of the VMNvl in each sex for analysis of glycogen synthase (GS) and glycogen phosphorylase (GP)­muscle type (GPmm; norepinephrine­sensitive) and GP­brain type (GPbb; glucoprivic­sensitive) isoform expression during and after IIH. Hypoglycemic suppression of VMNvl GS levels in males disappeared or continued after reestablishment of euglycemia, according to sampled segment. Yet, reductions in female VMNvl GS persisted after IIH. Males exhibited reductions in GPmm content in select rostro­caudal VMNvl segments, but this protein declined in each segment post­hypoglycemia. Females, rather, showed augmented or diminished GPmm levels during IIH, but no residual effects of IIH on this protein. In each sex, region­specific up­ or down­regulation of VMNvl GPbb profiles during glucose decrements were undetected post­recovery from IIH. Results provide novel proof of estradiol­dependent sex­dimorphic patterns of VMNvl GP variant expression at specific rostro­caudal levels of this critical gluco­regulatory structure. Sex differences in persistence of IIH­associated GS and GPmm patterns of expression after restoration of euglycemia infer that VMNvl recovery from this metabolic stress may involve dissimilar glycogen accumulation in male versus female.


Assuntos
Estradiol/farmacologia , Glicogênio Fosforilase/metabolismo , Hipoglicemiantes/farmacologia , Fatores Sexuais , Núcleo Hipotalâmico Ventromedial/efeitos dos fármacos , Animais , Estradiol/metabolismo , Feminino , Glucose/metabolismo , Glucose/farmacologia , Glicogênio/metabolismo , Glicogênio/farmacologia , Glicogênio Fosforilase/farmacologia , Masculino , Ratos
5.
Sci Total Environ ; 776: 145724, 2021 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-33652314

RESUMO

We made the first and successful attempt to detect SARS-CoV-2 genetic material in the vicinity wastewaters of an isolation centre i.e. Shaheed Bhulu Stadium, situated at Noakhali, Southeastern Bangladesh. Owing to the fact that isolation centre, in general, always contained a constant number of 200 COVID-19 patients, the prime objective of the study was to check if several drains carrying RNA of coronavirus are actually getting diluted or accumulated along with the sewage network. Our finding suggested that while the temporal variation of the genetic load decreased in small drains over the span of 50 days, the main sewer exhibited accumulation of SARS-CoV-2 RNA. Other interesting finding displays that probably distance of sampling location in meters is not likely to have a significant impact on the detected gene concentration, although the quantity of the RNA extracted in the downstream of the drain was higher. These findings are of immense value from the perspective of wastewater surveillance of COVID-19, as they largely imply that we do not need to monitor every wastewater system, and probably major drains monitoring may illustrate the city health. Perhaps, we are reporting the accumulation of SARS-CoV-2 genetic material along with the sewer network i.e. from primary to tertiary drains. The study sought further data collection in this line to simulate conditions prevailed in most of the developing countries and to shed further light on decay/accumulation processes of the genetic load of the SARS-COV-2.


Assuntos
COVID-19 , SARS-CoV-2 , Bangladesh , Cidades , Humanos , RNA Viral , Águas Residuárias
6.
AIMS Neurosci ; 8(1): 133-147, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33490375

RESUMO

Hypoglycemia activates the ultra-sensitive energy gauge 5'-AMP-activated protein kinase (AMPK) in ventromedial hypothalamic nucleus (VMN) gluco-regulatory neurons. The VMN is exemplified by high levels of expression of the enzyme aromatase, which converts testosterone to estradiol. This study examined the hypothesis that neuroestradiol imposes sex-dimorphic control of VMN AMPK activity during eu- and/or hypoglycemia. VMN tissue corresponding to distinct rostro-caudal segments was obtained by micropunch dissection from testes-intact male and estradiol-replaced ovariectomized female rats that were infused intracerebroventricularly with the aromatase inhibitor letrozole (Lz) before subcutaneous insulin (INS) injection. In euglycemic rats, Lz treatment elevated (male) or decreased (female) middle VMN phosphoAMPK content, with concurrent effects on total AMPK expression. Lz prevented hypoglycemic up-regulation of the mean pAMPK/AMPK ratio in rostral and middle segments of the male VMN, and significantly inhibited this proportion throughout the VMN of hypoglycemic female rats. Lz prevented glucagon secretion in hypoglycemic rats of each sex, and abolished hypoglycemic hypercorticosteronemia in males. Results show that neuroestradiol regulation of VMN AMPK activity during euglycemia is region-specific and gender-divergent, e.g. inhibitory in males versus stimulatory in females. Intra-VMN distribution of hypoglycemia-activated AMPK varies between sexes, but in each sex, locally-generated estradiol is critical for sensor reactivity to this stimulus. Coincident Lz attenuation of VMN AMPK and counter-regulatory hormone responses to hypoglycemia infers a possible cause-and-effect association. Further effort is needed to elucidate the cellular and molecular mechanisms that underlie sex-dimorphic neuroestradiol control of VMN total AMPK and phosphoAMPK expression during distinct metabolic states.

7.
J Mol Neurosci ; 71(5): 1082-1094, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33231812

RESUMO

The ventromedial hypothalamic nucleus-ventrolateral part (VMNvl) is an estradiol-sensitive structure that controls sex-specific behavior. Electrical reactivity of VMNvl neurons to hypoglycemia infers that cellular energy stability is monitored there. Current research investigated the hypothesis that estradiol elicits sex-dimorphic patterns of VMNvl metabolic sensor activation and gluco-regulatory neurotransmission during hypoglycemia. Rostral-, middle-, and caudal-VMNvl tissue was separately micropunch-dissected from letrozole (Lz)- or vehicle-injected male and estradiol- or vehicle-implanted ovariectomized (OVX) female rats for Western blot analysis of total and phosphorylated 5'-AMP-activated protein kinase (AMPK) protein expression and gluco-stimulatory [neuronal nitric oxide synthase (nNOS); steroidogenic factor-1 (SF1) or -inhibitory (glutamate decarboxylase65/67 (GAD)] transmitter marker proteins after sc insulin (INS) or vehicle injection. In both sexes, hypoglycemic up-regulation of phosphoAMPK was estradiol-dependent in rostral and middle, but not caudal VMNvl. AMPK activity remained elevated after recovery from hypoglycemia over the rostro-caudal VMNvl in female, but only in the rostral segment in male. In each sex, hypoglycemia correspondingly augmented or suppressed nNOS profiles in rostral and middle versus caudal VMNvl; these segmental responses persisted longer in female. Rostral and middle segment SF1 protein was inhibited by estradiol-independent mechanisms in hypoglycemic males, but increased by estradiol-reliant mechanisms in female. After INS injection, GAD expression was inhibited in the male rostral VMNvl without estradiol involvement, but this hormone was required for broader suppression of this profile in the female. Neuroanatomical variability of VMNvl metabolic transmitter reactivity to hypoglycemia underscores the existence of functionally different subgroups in that structure. The regional distribution and estradiol sensitivity of hypoglycemia-sensitive VMNvl neurons of each neurochemical phenotype evidently vary between sexes.


Assuntos
Estradiol/metabolismo , Glucose/metabolismo , Hipoglicemia/metabolismo , Hipotálamo/metabolismo , Proteínas Quinases/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Animais , Estradiol/farmacologia , Feminino , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Hipotálamo/efeitos dos fármacos , Insulina/metabolismo , Insulina/farmacologia , Masculino , Óxido Nítrico Sintase Tipo I/genética , Óxido Nítrico Sintase Tipo I/metabolismo , Proteínas Quinases/genética , Fatores de Processamento de RNA/genética , Fatores de Processamento de RNA/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
8.
BMC Neurosci ; 21(1): 51, 2020 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-33238883

RESUMO

BACKGROUND: Ventromedial hypothalamic nucleus (VMN) gluco-regulatory transmission is subject to sex-specific control by estradiol. The VMN is characterized by high levels of aromatase expression. METHODS: The aromatase inhibitor letrozole (LZ) was used with high-resolution microdissection/Western blot techniques to address the hypothesis that neuroestradiol exerts sex-dimorphic control of VMN neuronal nitric oxide synthase (nNOS) and glutamate decarboxylase65/67 (GAD) protein expression. Glycogen metabolism impacts VMN nNOS and GAD profiles; here, LZ treatment effects on VMN glycogen synthase (GS) and phosphorylase brain- (GPbb; glucoprivic-sensitive) and muscle (GPmm; norepinephrine-sensitive) variant proteins were examined. RESULTS: VMN aromatase protein content was similar between sexes. Intracerebroventricular LZ infusion of testes-intact male and ovariectomized, estradiol-replaced female rats blocked insulin-induced hypoglycemic (IIH) up-regulation of this profile. LZ exerted sex-contingent effects on basal VMN nNOS and GAD expression, but blocked IIH-induced NO stimulation and GAD suppression in each sex. Sex-contingent LZ effects on basal and hypoglycemic patterns of GPbb and GPmm expression occurred at distinctive levels of the VMN. LZ correspondingly down- or up-regulated baseline pyruvate recycling pathway marker protein expression in males (glutaminase) and females (malic enzyme-1), and altered INS effects on those proteins. CONCLUSIONS: Results infer that neuroestradiol is required in each sex for optimal VMN metabolic transmitter signaling of hypoglycemic energy deficiency. Sex differences in VMN GP variant protein levels and sensitivity to aromatase may correlate with sex-dimorphic glycogen mobilization during this metabolic stress. Neuroestradiol may also exert sex-specific effects on glucogenic amino acid energy yield by actions on distinctive enzyme targets in each sex.


Assuntos
Estradiol/fisiologia , Regulação da Expressão Gênica/genética , Glucose/metabolismo , Glicogênio/metabolismo , Caracteres Sexuais , Núcleo Hipotalâmico Ventromedial/metabolismo , Núcleo Hipotalâmico Ventromedial/fisiologia , Animais , Inibidores da Aromatase/farmacologia , Terapia de Reposição de Estrogênios , Feminino , Glutamato Descarboxilase/metabolismo , Glutaminase/metabolismo , Glicogênio Sintase/metabolismo , Letrozol/farmacologia , Malato Desidrogenase/metabolismo , Masculino , Óxido Nítrico Sintase/metabolismo , Ovariectomia , Ratos , Ratos Sprague-Dawley
9.
Neurosci Lett ; 737: 135284, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32853718

RESUMO

Estrogen receptors control hypothalamic astrocyte glycogen accumulation in vitro. Glycogen metabolism impacts metabolic transmitter signaling in the ventromedial hypothalamic nucleus (VMN), a key glucoregulatory structure. Aromatase, the enzyme that converts testosterone to estradiol, is expressed at high levels in the VMN. Here, the aromatase inhibitor letrozole (Lz) was used alongside high-resolution microdissection/UPHLC-electrospray ionization-mass spectrometric methods to determine if neuroestradiol imposes sex-specific control of VMN glycogen content during glucostasis and/or glucoprivation. Testes-intact male and estradiol-replaced ovariectomized female rats were pretreated by lateral ventricular letrozole (Lz) infusion prior to subcutaneous insulin (INS) injection. Vehicle-treated female controls exhibited higher VMN glycogen content compared to males. Lz increased VMN glycogen levels in males, not females. INS-induced hypoglycemia (IIH) elevated (males) or diminished (females) rostral VMN glycogen accumulation. Induction of IIH in Lz-pretreated animals reduced male VMN glycogen mass, but augmented content in females. Data provide novel evidence for regional variation, in both sexes, in glycogen reactivity to IIH. Results highlight sex-dimorphic neuroestradiol regulation of VMN glycogen amassment during glucostasis, e.g. inhibitory in males versus insignificant in females. Locally-generated estradiol is evidently involved in hypoglycemic enhancement of male VMN glycogen, but conversely limits glycogen content in hypoglycemic females. Further research is needed to characterize mechanisms that underlie the directional shift in aromatase regulation of VMN glycogen in eu- versus hypoglycemic male rats and gain in negative impact in hypoglycemic females.


Assuntos
Aromatase/metabolismo , Glicemia/metabolismo , Glicogênio/metabolismo , Hipoglicemia/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , Animais , Inibidores da Aromatase/farmacologia , Feminino , Hipoglicemia/induzido quimicamente , Insulina , Letrozol/farmacologia , Masculino , Ratos , Ratos Sprague-Dawley , Fatores Sexuais , Núcleo Hipotalâmico Ventromedial/efeitos dos fármacos
10.
Mol Cell Biochem ; 473(1-2): 39-50, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32779041

RESUMO

Hypoglycemia is a detrimental complication of rigorous management of type 1 diabetes mellitus. Moderate hypoglycemia (MH) preconditioning of male rats partially affords protection from loss of vulnerable brain neurons to severe hypoglycemia (SH). Current research investigated whether MH preconditioning exerts sex-dimorphic effects on hippocampal CA1 neuron bio-energetic and anti-oxidant responses to SH. SH up-regulated CA1 glucose or monocarboxylate transporter proteins in corresponding hypoglycemia-naïve male versus female rats; precedent MH amplified glucose transporter expression in SH irrespective of sex. Sex-differentiating SH effects on glycolytic and tricarboxylic pathway markers correlated with elevated tissue ATP content and diminished CA1 5'-AMP-activated protein kinase (AMPK) activation in females. MH-preconditioned suppression of mitochondrial energy pathway enzyme profiles and tissue ATP in SH rats coincided with amplified CA1 AMPK activity in both sexes. Anti-oxidative stress enzyme protein responses to SH were primarily sex-contingent; preconditioning amplified most of these profiles, yet exacerbated expression of lipid and protein oxidation markers in SH male and female rats, respectively. Results show that MH preconditioning abolishes female CA1 neuron neuroprotection of positive energy balance through SH, resulting in augmented CA1 AMPK activity and oxidative injury and diminished tissue ATP in hypoglycemia-conditioned versus naïve rats in each sex. It is unclear if SH elicits differential rates of CA1 neuronal destruction in the two sexes, or how MH may impact sex-specific cell loss. Further research is needed to determine if molecular mechanism(s) that maintain female CA1 neuron metabolic stability in the absence of MH preconditioning can be leveraged for therapeutic prevention of hypoglycemic nerve cell damage.


Assuntos
Região CA1 Hipocampal/metabolismo , Glicólise , Hipoglicemia/metabolismo , Neurônios/metabolismo , Caracteres Sexuais , Proteínas Quinases Ativadas por AMP/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Região CA1 Hipocampal/patologia , Feminino , Hipoglicemia/patologia , Masculino , Neurônios/patologia , Oxirredução , Ratos , Ratos Sprague-Dawley
11.
Int J Mol Sci ; 21(6)2020 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-32188013

RESUMO

The mediobasal hypothalamus (MBH) shapes the neural regulation of glucostasis by 5'-AMP-activated protein kinase (AMPK)-dependent mechanisms. Yet, the neurochemical identity and neuroanatomical distribution of MBH neurons that express glucoprivic-sensitive AMPK remain unclear. The neurotransmitters γ-aminobutyric acid (GABA) and nitric oxide (NO) act within the MBH to correspondingly inhibit or stimulate glucose counter-regulation. The current review highlights recent findings that GABA and NO, neurons located in the ventromedial hypothalamic nucleus (VMN), a distinct important element of the MBH, are direct targets of noradrenergic regulatory signaling, and thereby, likely operate under the control of hindbrain metabolic-sensory neurons. The ovarian hormone estradiol acts within the VMN to govern energy homeostasis. Discussed here is current evidence that estradiol regulates GABA and NO nerve cell receptivity to norepinephrine and moreover, controls the noradrenergic regulation of AMPK activity in each cell type. Future gains in insight on mechanisms underpinning estradiol's impact on neurotransmitter communication between the hindbrain and hypothalamic AMPKergic neurons are expected to disclose viable new molecular targets for the therapeutic simulation of hormonal enhancement of neuro-metabolic stability during circumstances of diminished endogenous estrogen secretion or glucose dysregulation.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Estradiol/farmacologia , Norepinefrina/metabolismo , Células Receptoras Sensoriais/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , Animais , Glicemia/metabolismo , Feminino , Glucose/metabolismo , Glutamato Descarboxilase , Glicogênio/metabolismo , Homeostase , Hipotálamo , Óxido Nítrico , Óxido Nítrico Sintase , Receptores de Estrogênio , Rombencéfalo , Transativadores , Ácido gama-Aminobutírico
12.
Neuroscience ; 409: 253-260, 2019 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-30954669

RESUMO

Estrogen receptor-alpha (ERα) and -beta (ERß) occur in key elements of the brain gluco-homeostatic network in both sexes, including the hindbrain dorsal vagal complex (DVC), but the influence of distinct receptor populations on this critical function is unclear. The ventromedial hypothalamic nucleus (VMN) maintains glucose balance by integrating nutrient, endocrine, and neurochemical cues, including metabolic sensory information supplied by DVC A2 noradrenergic neurons. Current research utilized the selective ERα and ERß antagonists MPP and PHTPP to characterize effects of DVC ERs on VMN norepinephrine (NE) activity and metabolic neurotransmitter signaling in insulin-induced hypoglycemic (IIH) male rats. Data show that ERß inhibits VMN glycogen synthase and stimulates phosphorylase protein expression, while attenuating hypoglycemic augmentation of glycogen content. Furthermore, both ERs attenuate VMN glucose concentrations during IIH. Hypoglycemic up-regulation of nitric oxide (NO) and brain-derived neurotrophic factor (BDNF) signaling was correspondingly driven by ERα or -ß, whereas GABA and steroidogenic factor-1 were respectively suppressed independently of ER input or by ERß. IIH intensified VMN NE accumulation by ERß-dependent mechanisms, but did not alter NE levels in other gluco-regulatory loci. ERß amplified the magnitude of insulin-induced decline in blood glucose. Both ERs regulate corticosterone, but not glucagon secretion during IIH and oppose hypoglycemic diminution of circulating free fatty acids. These findings identify distinguishing versus common VMN functions targeted by DVC ERα and -ß. Sex differences in hypoglycemic VMN NE accumulation, glycogen metabolism, and transmitter signaling may involve, in part, discrepant regulatory involvement or differential magnitude of impact of these hindbrain ERs.


Assuntos
Glucose/metabolismo , Glicogênio/metabolismo , Hipoglicemia/metabolismo , Receptores de Estrogênio/metabolismo , Rombencéfalo/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Corticosterona/metabolismo , Masculino , Óxido Nítrico/metabolismo , Norepinefrina/metabolismo , Piperidinas/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Estrogênio/antagonistas & inibidores , Rombencéfalo/efeitos dos fármacos , Núcleo Hipotalâmico Ventromedial/efeitos dos fármacos
13.
Neuroscience ; 379: 103-114, 2018 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-29534973

RESUMO

The hypothalamic energy sensor adenosine 5'-monophosphate-activated protein kinase (AMPK), an important regulator of counter-regulatory responses to hypoglycemia, responds to pharmacological manipulation of hindbrain AMPK activity. Dorsomedial hindbrain A2 noradrenergic neurons express hypoglycemia-sensitive metabolo-sensory biomarkers, including AMPK. Here, adult male rats were pretreated by intra-caudal fourth ventricular administration of the selective neurotoxin 6-hydroxydopamine (6-OHDA) to determine if catecholamine signaling from the aforesaid site governs hypothalamic AMPK activation during insulin-induced hypoglycemia (IIH). Micropunched arcuate (ARH), ventromedial (VMH), paraventricular (PVH), dorsomedial (DMH) nuclei and lateral hypothalamic area (LHA) tissues were obtained at the neutral protamine Hagedorn insulin-induced hypoglycemic nadir, coincident with A2 AMPK activation, for Western blot analysis of AMPK, phospho-AMPK (pAMPK), and relevant metabolic neuropeptides. ARH, VMH, LHA, and DMH norepinephrine levels were altered according to insulin dose; 6-OHDA-mediated reversal of these responses was site-specific. IIH elevated LHA and reduced VMH pAMPK protein, profiles that were respectively unchanged or increased by 6-OHDA. PVH and ARH pAMPK was resistant to IIH, but augmented in ARH of neurotoxin- plus insulin-treated rats. ARH neuropeptide Y (NPY) and pro-opiomelanocortin (POMC) proteins were correspondingly increased or refractory to IIH; 6-OHDA pretreatment normalized NPY and elevated POMC expression after insulin injection. Results demonstrate site-specific bi-directional adjustments in hypothalamic AMPK reactivity to hypoglycemia. Intensification of ARH/VMH pAMPK by 6-OHDA implies dorsomedial hindbrain improvement of energy balance in those sites during IIH. Neurotoxin-mediated augmentation versus suppression of basal catabolic (ARH POMC/VMH steroidogenic factor-1) or IIH-associated anabolic (ARH NPY) neuropeptide profiles, respectively, may involve local AMPK-dependent against independent mechanisms.


Assuntos
Adenilato Quinase/metabolismo , Catecolaminas/metabolismo , Hipoglicemia/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Rombencéfalo/metabolismo , Adrenérgicos/farmacologia , Animais , Hipotálamo/efeitos dos fármacos , Insulina/administração & dosagem , Masculino , Neuropeptídeo Y/metabolismo , Oxidopamina/farmacologia , Fosforilação , Pró-Opiomelanocortina/metabolismo , Ratos Sprague-Dawley , Rombencéfalo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
14.
Eur J Nutr ; 53(8): 1699-706, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24573416

RESUMO

PURPOSE: To investigate the presence of anti-angiotensin converting enzyme (ACE) factors in aqueous extract of tomato. METHODS: The bio-guided fractionation of the aqueous extract of tomato produced a sugar-free, heat-stable fraction with molecular mass <1,000 Da from tomatoes. The sugar-free tomato extract (TE) was tested for its anti-ACE activity using human plasma and rabbit lung pure ACE. In addition, its effect on human platelet aggregation induced by ADP, collagen or arachidonic acid was determined. The mechanism of platelet inhibitory action of TE was investigated by measuring platelet factor 4 (PF4) release and cAMP synthesis by platelets. RESULTS: Typically, 100 g tomatoes produced 72.2 ± 4.7 mg of TE. This extract inhibited both platelet aggregation and plasma ACE activity in a dose-dependent manner. It inhibited platelet aggregation in response to ADP, collagen or arachidonic acid, and inhibitory action was mediated in part by reducing platelet PF4 release and by stimulating cAMP synthesis. The IC50 value of TE for ADP-induced platelet aggregation was 0.4 ± 0.02 mg/ml, whereas the IC50 value for ACE enzyme inhibition was 1.40 ± 0.04 mg/ml. Both the TE and commercially available sugar-free TE, Fruitflow(®)-2 had similar amount of catechin, and also had equal inhibitory potencies against platelet aggregation and plasma ACE activity. CONCLUSION: Together these data indicate that aqueous extract of tomatoes contain anti-ACE factors in addition to previously described anti-platelet factors.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Peptidil Dipeptidase A/sangue , Extratos Vegetais/farmacologia , Solanum lycopersicum/química , Animais , Ácido Araquidônico/metabolismo , Colágeno/metabolismo , AMP Cíclico/metabolismo , Hidroxibenzoatos/análise , Hidroxibenzoatos/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Coelhos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...