Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
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.
Neuropeptides ; 99: 102324, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36791640

RESUMO

BACKGROUND: The oxidizable glycolytic end-product L-lactate is a gauge of nerve cell metabolic fuel stability that metabolic-sensory hindbrain A2 noradrenergic neurons impart to the brain glucose-regulatory network. Current research investigated the premise that hindbrain lactate deficiency exerts sex-specific control of energy sensor and transmitter marker protein responses to hypoglycemia in ventromedial hypothalamic nucleus (VMN) glucose-regulatory nitrergic and γ-aminobutyric acid (GABA) neurons. METHODS: Nitric oxide synthase (nNOS)- or glutamate decarboxylase65/67 (GAD)-immunoreactive neurons were laser-catapult-microdissected from male and female rat VMN after subcutaneous insulin injection and caudal fourth ventricular L-lactate or vehicle infusion for Western blot protein analysis. RESULTS: Hindbrain lactate repletion reversed hypoglycemia-associated augmentation (males) or inhibition (females) of nitrergic neuron nNOS expression, and prevented up-regulation of phosphorylated AMPK 5'-AMP-activated protein kinase (pAMPK) expression in those neurons. Hypoglycemic suppression of GABAergic neuron GAD protein was averted by exogenous lactate over the rostro-caudal length of the male VMN and in the middle region of the female VMN. Lactate normalized GABA neuron pAMPK profiles in hypoglycemic male (caudal VMN) and female (all VMN segments) rats. Hypoglycemic patterns of norepinephrine (NE) signaling were lactate-dependent throughout the male VMN, but confined to the rostral and middle female VMN. CONCLUSIONS: Results document, in each sex, regional VMN glucose-regulatory transmitter responses to hypoglycemia that are controlled by hindbrain lactate status. Hindbrain metabolic-sensory regulation of hypoglycemia-correlated nitric oxide or GABA release may entail AMPK-dependent mechanisms in specific VMN rostro-caudal segments in each sex. Additional effort is required to examine the role of hindbrain lactoprivic-sensitive VMN neurotransmitters in lactate-mediated attenuation of hypoglycemic hyperglucagonemia and hypercorticosteronemia in male and female rats.


Assuntos
Neurônios Adrenérgicos , Hipoglicemia , Ratos , Feminino , Masculino , Animais , Núcleo Hipotalâmico Ventromedial/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Ácido Láctico , Ratos Sprague-Dawley , Glucose/metabolismo , Hipoglicemia/metabolismo , Rombencéfalo/metabolismo , Norepinefrina/metabolismo , Hipoglicemiantes , Neurônios Adrenérgicos/metabolismo
3.
J Chem Neuroanat ; 124: 102132, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35772680

RESUMO

The inhibitory amino acid transmitter γ-aminobutryic acid (GABA) acts within the ventromedial hypothalamus to regulate systemic glucose homeostasis, but the issue of whether this neurochemical signal originates locally or is supplied by afferent innervation remains controversial. Here, combinatory in situ immunocytochemistry/laser-catapult microdissection/single-cell multiplex qPCR techniques were used to investigate the premise that ventromedial hypothalamic nucleus ventrolateral (VMNvl) and/or dorsomedial (VMNdm) division neurons contain mRNAs that encode glutamate decarboxylase (GAD)65 or GAD67 and metabolic-sensory biomarkers, and that expression of these genes is sex-dimorphic. In male and female rats, GAD65 mRNA was elevated in VMNvl versus VMNdm GAD65/67-immunopositive (-ir) neurons, yet the female exhibited higher GAD67 transcript content in VMNdm versus VMNvl GABAergic nerve cells. Estrogen receptor (ER)-alpha transcripts were lower in female versus male GABA neurons from either VMN division; ER-beta and G-protein-coupled ER-1 mRNA expression profiles were also comparatively reduced in cells from female versus male VMNvl. VMNvl and VMNdm GAD65/67-ir-positive neurons showed equivalent levels of glucokinase and sulfonylurea receptor-1 mRNA between sexes. 5'-AMP-activated protein kinase-alpha 1 (AMPKα1) and -alpha 2 (AMPKα2) transcripts were lower in female versus male VMNdm GABAergic neurons, yet AMPKα2 mRNA levels were higher in cells acquired from female versus male VMNvl. Current studies document GAD65 and -67 gene expression in VMNvl and VMNdm GAD65/67-ir-positive neurons in each sex. Results infer that GABAergic neurons in each division may exhibit sex differences in receptiveness to estradiol. Outcomes also support the prospect that energy sensory function by this neurotransmitter cell type may predominate in the VMNvl in female versus VMNdm in the male.


Assuntos
Proteínas Quinases Ativadas por AMP , Núcleo Hipotalâmico Ventromedial , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Receptor alfa de Estrogênio , Feminino , Neurônios GABAérgicos/metabolismo , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Masculino , RNA Mensageiro/metabolismo , Ratos , Receptores de Estrogênio/metabolismo
4.
J Chem Neuroanat ; 122: 102102, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35483611

RESUMO

Caudal hindbrain A2 noradrenergic neurons provide critical metabolic-sensory input to the brain glucostatic circuitry. In males, insulin-induced hypoglycemia (IIH)-associated patterns of A2 cell dopamine-beta-hydroxylase (DßH) protein expression reflect diminution of the oxidizable fuel L-lactate, yet DßH exhibits sex-dimorphic responses to IIH. Here, retrograde tracing and combinatory single-cell laser-microdissection/multiplex qPCR techniques were used to examine whether lactate imposes sex-specific control of hypoglycemia-associated metabolic-sensory function and noradrenergic neurotransmission in A2 neurons that innervate the ventromedial hypothalamic nucleus (VMN), a key glucose-regulatory structure. VMN-projecting A2 neurons from each sex were characterized by presence or absence of nuclear glucokinase regulatory protein (nGKRP) immunoreactivity (-ir). IIH caused lactate-reversible up- or down-regulation of DßH mRNA in male and female nGKRP-ir-positive A2 neurons, respectively, and stimulated glucokinase (GCK) and sulfonylurea receptor-1 (SUR-1) gene expression in these cells in each sex. Hypoglycemia did not alter DßH, GCK, and SUR-1 transcript profiles in nGKRP-ir-negative male or female A2 neurons innervating the VMN. Estrogen receptor (ER) gene profiles in nGKRP-ir-positive neurons showed sex-specific [ER-alpha; G-protein-coupled estrogen-receptor-1 (GPER)] or sex-monomorphic (ER-beta) transcriptional responses to IIH. Fewer ER gene profiles were affected by IIH in nGKRP-ir-negative A2 neurons from male or female rats. Results show that during IIH, VMN-projecting A2 neurons may deliver altered, sex-dependent (nGKRP-positive) or unaffected (nGKRP-negative) noradrenergic input to the VMN. In each sex, metabolic-sensory gene profiles were reactive to hypoglycemia in nGKRP-ir-positive, not -negative A2 cells. Further studies are needed to elucidate the role of GKRP in transduction of metabolic imbalance into noradrenergic signaling, and to determine if input by one or more ER variants establishes sex differences in DßH transcriptional sensitivity to IIH.


Assuntos
Neurônios Adrenérgicos , Hipoglicemia , Neurônios Adrenérgicos/metabolismo , Animais , Biomarcadores/metabolismo , Catecolaminas/metabolismo , Feminino , Expressão Gênica , Hipoglicemia/genética , Hipoglicemia/metabolismo , Ácido Láctico/metabolismo , Ácido Láctico/farmacologia , Masculino , Ratos , Rombencéfalo/metabolismo , Núcleo Hipotalâmico Ventromedial
5.
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
6.
Brain Struct Funct ; 226(4): 1053-1065, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33580322

RESUMO

Recurring insulin-induced hypoglycemia (RIIH) in males correlates with maladaptive glucose counter-regulatory collapse and acclimated expression of ventromedial hypothalamic nucleus (VMN) nitric oxide (NO) and γ-aminobutyric acid (GABA) metabolic transmitter biomarkers, e.g., neuronal nitric oxide synthase (nNOS) and glutamate decarboxylase65/67 (GAD). Hindbrain noradrenergic neurons innervate the VMN, where norepinephrine regulates nNOS and GAD expression. Current research investigated the hypothesis that antecedent hypoglycemia (AH) exposure causes sex-dimorphic habituation of VMN glucoregulatory biomarker proteins between and/or during serial hypoglycemic bouts, and that hindbrain catecholaminergic (CA) signaling may control sex-specific adaptation of one or more of these proteins. Data show that upon recovery from AH, females exhibit CA-mediated reductions in baseline VMN nNOS, GAD, steroidogenic factor-1 (SF-1), and brain-derived neurotrophic factor (BNDF) expression compared to euglycemic profiles. In males, however, AH caused 6-OHDA-insensitive suppression of only basal SF-1 levels in the VMN. VMN transmitter protein acclimation to RIIH was sex-contingent, as differential nNOS, GAD, SF-1, and BDNF responses to a single vs final bout of hypoglycemia occur in males, whereas females show acclimated reactivity of GAD and SF-1 only to renewed hypoglycemia. CA-mediated and -independent habituation of distinctive VMN protein profiles occurred in each sex. Further research is necessary to evaluate, in each sex, effects of altered baseline VMN metabolic neurotransmitter signals on glucose homeostasis as well as non-metabolic functions under the control of those neurochemicals. It would also be insightful to learn if and how sex-contingent habituation of VMN transmitter responses to hypoglycemia contribute to sex-dimorphic patterns of glucose counter-regulation during RIIH.


Assuntos
Hipoglicemia , Núcleo Hipotalâmico Ventromedial , Animais , Biomarcadores , Feminino , Glucose , Glicogênio/metabolismo , Hipoglicemia/induzido quimicamente , Insulinas , Masculino , Ratos , Ratos Sprague-Dawley , Caracteres Sexuais , Núcleo Hipotalâmico Ventromedial/metabolismo
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.
J Chem Neuroanat ; 109: 101845, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32599255

RESUMO

Hindbrain estrogen receptors (ER) impose sex-dimorphic control of counter-regulatory hormone and hypothalamic glucoregulatory transmitter and glycogen metabolic responses to hypoglycemia. A2 noradrenergic neurons are estradiol- and metabolic-sensitive. Estradiol controls dopamine-beta-hydroxylase (DBH) protein habituation to recurrent insulin-induced hypoglycemia (RIIH) in females. Current research investigated the premise that sex-dimorphic patterns of A2 ER variant acclimation to RIIH correlate with differential A2 DBH and 5'-AMP-activated protein kinase (AMPK) adaptation to RIIH. A2 neurons were laser-catapult-microdissected from male and female rats after one or four insulin injections for Western blot analysis. A2 pAMPK and DBH levels were increased in males, but suppressed in females after single insulin dosing. ER-alpha (ERα) and -beta (ERß) protein profiles were unaffected or decreased by acute hypoglycemia in each sex, whereas G protein-linked ER-1 (GPER) reactivity varied by sex. Antecedent hypoglycemia diminished basal A2 ERα/GPER and elevated ERß content in each sex, yet reduced pAMPK and DBH levels in female rats only. Reintroduced hypoglycemia suppressed A2 ERß levels in each sex, but altered DBH (↓), ERα (↓), and GPER (↑) levels in males only. Data document sex differences in A2 DBH adaptation to RIIH, e.g. a shift from positive-to-negative response in males versus loss of negative reactivity in females, as well as attenuated AMPK activation in both sexes. Between hypoglycemic episodes, A2 neurons in each sex likely exhibit diminished sensitivity to ERα/GPER signaling, but heightened receptivity to ERß input. RIIH-induced changes in ERα and GPER expression in male but not female may contribute to DBH suppression (males) versus no change (females) relative to adapted baseline expression.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Neurônios Adrenérgicos/metabolismo , Dopamina beta-Hidroxilase/metabolismo , Hipoglicemia/metabolismo , Insulina/farmacologia , Receptores de Estrogênio/metabolismo , Neurônios Adrenérgicos/efeitos dos fármacos , Animais , Feminino , Masculino , Ratos , Ratos Sprague-Dawley , Fatores Sexuais
10.
Neuroscience ; 411: 211-221, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31085279

RESUMO

Neural substrates for estrogen regulation of glucose homeostasis remain unclear. Female rat dorsal vagal complex (DVC) A2 noradrenergic neurons are estrogen- and metabolic-sensitive. The ventromedial hypothalamic nucleus (VMN) is a key component of the brain network that governs counter-regulatory responses to insulin-induced hypoglycemia (IIH). Here, the selective estrogen receptor-alpha (ERα) or -beta (ERß) antagonists MPP and PHTPP were administered separately to the caudal fourth ventricle to address the premise that these hindbrain ER variants exert distinctive control of VMN reactivity to IIH in the female sex. Data show that ERα governs hypoglycemic patterns of VMN astrocyte glycogen metabolic enzyme, e.g. glycogen synthase and phosphorylase protein expression, whereas ERß mediates local glycogen breakdown. DVC ERs also regulate VMN neurotransmitter signaling of energy sufficiency [γ-aminobutyric acid] or deficiency [nitric oxide, steroidogenic factor-1] during IIH. Neither hindbrain ER mediates IIH-associated diminution of VMN norepinephrine (NE) content. Both ERs oppose hypoglycemic hyperglucagonemia, while ERß contributes to reduced corticosterone output. Outcomes reveal that input from the female hindbrain to the VMN is critical for energy reserve mobilization, metabolic transmitter signaling, and counter-regulatory hormone secretion during hypoglycemia, and that ERs control those cues. Evidence that VMN NE content is not controlled by hindbrain ERα or -ß implies that these receptors may regulate VMN function via NE-independent mechanisms, or alternatively, that other neurotransmitter signals to the VMN may control local substrate receptivity to NE.


Assuntos
Glicogênio/metabolismo , Hipoglicemia/metabolismo , Receptores de Estrogênio/metabolismo , Rombencéfalo/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Antagonistas do Receptor de Estrogênio/farmacologia , Feminino , Óxido Nítrico Sintase Tipo I/metabolismo , Piperidinas/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Ratos , Rombencéfalo/efeitos dos fármacos , Fator Esteroidogênico 1/metabolismo , Núcleo Hipotalâmico Ventromedial/efeitos dos fármacos
11.
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
12.
Environ Sci Pollut Res Int ; 25(4): 3193-3199, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28631127

RESUMO

The objective of the present study was to elucidate the effects of cypermethrin on the embryo and the larvae of Gangetic mystus, Mystus cavasius. Therefore, fertilized eggs (n = 100) and 1-day-old larvae (n = 100) were exposed to six different concentrations of cypermethrin (0, 2, 4, 8, 16 and 32 µg L-1) in each of the 18 plastic bowls. Each of the treatment and control was maintained in three replicates. The LC10 and LC50 values for Gangetic mystus embryos and larvae were calculated using probit analysis. Results showed the mortality of embryos significantly increased with increasing cypermethrin concentrations. The 24-h LC10 and LC50 (with 95% confidence interval) values of cypermethrin for embryo were 0.42 (0.14-0.81) and 5.60 (4.16-7.19) µg L-1, respectively. Hatching success decreased and mortality of larvae increased significantly with increasing cypermethrin concentrations. The 24-h LC10 and LC50 values (with 95% confidence limits) of cypermethrin for larvae were 1.72 (1.24-2.20) and 11.57 (10.09-13.42) µg L-1, respectively; the 48-h LC10 and LC50 for larvae were 1.34 (0.83-1.89) and 8.25 (6.87-9.91) µg L-1, respectively; the 72-h LC10 and LC50 for larvae were 1.13 (0.63-1.66) and 6.12 (4.91-7.47) µg L-1, respectively. Furthermore, results showed several malformations in embryos and larvae when exposed to the two highest concentrations of cypermethrin. The findings of the study suggest that 2 µg L-1 cypermethrin concentration in the aquatic environment may have deleterious effects on the development and the reproduction of Gangetic mystus.


Assuntos
Peixes-Gato/crescimento & desenvolvimento , Embrião não Mamífero/efeitos dos fármacos , Inseticidas/toxicidade , Piretrinas/toxicidade , Animais , Relação Dose-Resposta a Droga , Dose Letal Mediana , Testes de Toxicidade Aguda
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA