Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cureus ; 16(2): e53921, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38465089

RESUMO

This case report delineates the occurrence and management of type 2 myocardial infarction (MI) in an 89-year-old woman following transcatheter aortic valve implantation (TAVI). The patient, with a history of severe aortic stenosis, hypertension, dyslipidemia, and colorectal cancer, presented with nausea and significant hypotension. Initial assessments revealed elevated troponin levels, atrial fibrillation, and ST-segment depression, leading to a diagnosis of type 2 MI. This condition was attributed to the interplay between left ventricular hypertrophy, hypotension-induced dehydration, and increased myocardial oxygen demand. The patient with post-TAVI exhibited dynamic changes in cardiac hemodynamics, with improvements in left ventricular function but persistent hypertrophy and diastolic dysfunction. This state, combined with hypotension due to diuretic-induced dehydration and atrial fibrillation, precipitated a mismatch in myocardial oxygen supply and demand. The cessation of diuretics and initiation of rehydration therapy stabilized her condition, with subsequent normalization of troponin levels and blood pressure. This case highlights the complexity of managing type 2 MI in elderly patients post-TAVI. It underscores the importance of holistic consideration of both myocardial oxygen supply and demand factors, particularly in left ventricular hypertrophy and diastolic dysfunction. The multifactorial nature of type 2 MI necessitates a tailored approach to diagnosis and management, emphasizing the need for comprehensive post-procedural care in patients undergoing TAVI.

2.
Sci Rep ; 13(1): 20495, 2023 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-37993510

RESUMO

The gonadotropin-releasing hormone (GnRH) pulse and surge are considered to be generated by arcuate kisspeptin/neurokinin B/dynorphin A (KNDy) neurons and anteroventral periventricular nucleus (AVPV) kisspeptin neurons, respectively, in female rodents. The majority of KNDy and AVPV kisspeptin neurons express κ-opioid receptors (KORs, encoded by Oprk1) in female rodents. Thus, this study aimed to investigate the effect of a conditional Oprk1-dependent Kiss1 deletion in kisspeptin neurons on the luteinizing hormone (LH) pulse/surge and fertility using Kiss1-floxed/Oprk1-Cre rats, in which Kiss1 was deleted in cells expressing or once expressed the Oprk1/Cre. The Kiss1-floxed/Oprk1-Cre female rats, with Kiss1 deleted in a majority of KNDy neurons, showed normal puberty while having a one-day longer estrous cycle and fewer pups than Kiss1-floxed controls. Notably, ovariectomized (OVX) Kiss1-floxed/Oprk1-Cre rats showed profound disruption of LH pulses in the presence of a diestrous level of estrogen but showed apparent LH pulses without estrogen treatment. Furthermore, Kiss1-floxed/Oprk1-Cre rats, with Kiss1 deleted in approximately half of AVPV kisspeptin neurons, showed a lower peak of the estrogen-induced LH surge than controls. These results suggest that arcuate and AVPV kisspeptin neurons expressing or having expressed Oprk1 have a role in maintaining normal GnRH pulse and surge generation, the normal length of the estrous cycle, and the normal offspring number in female rats.


Assuntos
Kisspeptinas , Hormônio Luteinizante , Ratos , Feminino , Animais , Kisspeptinas/metabolismo , Hormônio Luteinizante/farmacologia , Estrogênios/farmacologia , Hormônio Liberador de Gonadotropina/metabolismo , Neurocinina B/genética , Neurocinina B/metabolismo , Dinorfinas/metabolismo , Neurônios/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo
3.
J Reprod Dev ; 69(5): 227-238, 2023 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-37518187

RESUMO

Hypothalamic kisspeptin neurons are master regulators of mammalian reproduction via direct stimulation of gonadotropin-releasing hormone and consequent gonadotropin release. Here, we generated novel Kiss1 (kisspeptin gene)-Cre rats and investigated the developmental changes and sex differences in visualized Kiss1 neurons of Kiss1-Cre-activated tdTomato reporter rats. First, we validated Kiss1-Cre rats by generating Kiss1-expressing cell-specific Kiss1 knockout (Kiss1-KpKO) rats, which were obtained by crossing the current Kiss1-Cre rats with Kiss1-floxed rats. The resulting male Kiss1-KpKO rats lacked Kiss1 expression in the brain and exhibited hypogonadotropic hypogonadism, similar to the hypogonadal phenotype of global Kiss1 KO rats. Histological analysis of Kiss1 neurons in Kiss1-Cre-activated tdTomato reporter rats revealed that tdTomato signals in the anteroventral periventricular nucleus (AVPV) and arcuate nucleus (ARC) were not affected by estrogen, and that tdTomato signals in the ARC, AVPV, and medial amygdala (MeA) were sexually dimorphic. Notably, neonatal AVPV tdTomato signals were detected only in males, but a larger number of tdTomato-expressing cells were detected in the AVPV and ARC, and a smaller number of cells in the MeA was detected in females than in males at postpuberty. These findings suggest that Kiss1-visualized rats can be used to examine the effect of estrogen feedback mechanisms on Kiss1 expression in the AVPV and ARC. Moreover, the Kiss1-Cre and Kiss1-visualized rats could be valuable tools for further detailed analyses of sexual differentiation in the brain and the physiological role of kisspeptin neurons across the brain in rats.


Assuntos
Kisspeptinas , Caracteres Sexuais , Ratos , Animais , Feminino , Masculino , Kisspeptinas/metabolismo , Núcleo Arqueado do Hipotálamo/metabolismo , Estrogênios/metabolismo , Neurônios/metabolismo , Mamíferos/metabolismo
4.
J Reprod Dev ; 69(4): 192-197, 2023 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-37331801

RESUMO

Gonadal function is often suppressed during lactation in mammals including rodents, ruminants, and primates. This suppression is thought to be mostly due to the inhibition of the tonic (pulsatile) release of gonadotropin-releasing hormone (GnRH) and consequent gonadotropin. Accumulating evidence suggests that kisspeptin neurons in the arcuate nucleus (ARC) play a critical role in the regulation of pulsatile GnRH/gonadotropin release, and kisspeptin mRNA (Kiss1) and/or kisspeptin expression in the ARC are strongly suppressed by the suckling stimuli in lactating rats. This study aimed to examine whether the central enkephalin-δ-opioid receptor (DOR) signaling mediates the suckling-induced suppression of luteinizing hormone (LH) release in lactating rats. Central administration of a selective DOR antagonist increased the mean plasma LH levels and baseline of LH pulses in ovariectomized lactating mother rats compared to vehicle-injected control dams on day 8 of lactation without affecting the number of Kiss1-expressing cells and the intensity of Kiss1 mRNA signals in the ARC. Furthermore, the suckling stimuli significantly increased the number of enkephalin mRNA (Penk)-expressing cells and the intensity of Penk mRNA signals in the ARC compared to non-lactating control rats. Collectively, these results suggest that central DOR signaling, at least in part, mediates the suppression of LH release induced by suckling stimuli in lactating rats via indirect and/or direct inhibition of ARC kisspeptin neurons.


Assuntos
Kisspeptinas , Receptores Opioides delta , Feminino , Ratos , Animais , Kisspeptinas/genética , Lactação , Hormônio Luteinizante , Hormônio Liberador de Gonadotropina , Mamíferos
5.
J Neurosci ; 43(12): 2140-2152, 2023 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-36813577

RESUMO

Ovulation disorders are a serious problem for humans and livestock. In female rodents, kisspeptin neurons in the anteroventral periventricular nucleus (AVPV) are responsible for generating a luteinizing hormone (LH) surge and consequent ovulation. Here, we report that adenosine 5-triphosphate (ATP), a purinergic receptor ligand, is a possible neurotransmitter that stimulates AVPV kisspeptin neurons to induce an LH surge and consequent ovulation in rodents. Administration of an ATP receptor antagonist (PPADS) into the AVPV blocked the LH surge in ovariectomized (OVX) rats treated with a proestrous level of estrogen (OVX + high E2) and significantly reduced the ovulation rate in proestrous ovary-intact rats. AVPV ATP administration induced a surge-like LH increase in OVX + high E2 rats in the morning. Importantly, AVPV ATP administration could not induce the LH increase in Kiss1 KO rats. Furthermore, ATP significantly increased intracellular Ca2+ levels in immortalized kisspeptin neuronal cell line, and coadministration of PPADS blocked the ATP-induced Ca2+ increase. Histologic analysis revealed that the proestrous level of estrogen significantly increased the number of P2X2 receptor (an ATP receptor)-immunopositive AVPV kisspeptin neurons visualized by tdTomato in Kiss1-tdTomato rats. The proestrous level of estrogen significantly increased varicosity-like vesicular nucleotide transporter (a purinergic marker)-immunopositive fibers projecting to the vicinity of AVPV kisspeptin neurons. Furthermore, we found that some hindbrain vesicular nucleotide transporter-positive neurons projected to the AVPV and expressed estrogen receptor α, and the neurons were activated by the high E2 treatment. These results suggest that hindbrain ATP-purinergic signaling triggers ovulation via activation of AVPV kisspeptin neurons.SIGNIFICANCE STATEMENT Ovulation disorders, which cause infertility and low pregnancy rates, are a serious problem for humans and livestock. The present study provides evidence that adenosine 5-triphosphate, acting as a neurotransmitter in the brain, stimulates kisspeptin neurons in the anteroventral periventricular nucleus, known as the gonadotropin-releasing hormone surge generator, via purinergic receptors to induce the gonadotropin-releasing hormone/luteinizing hormone surge and ovulation in rats. In addition, histologic analyses indicate that adenosine 5-triphosphate is likely to be originated from the purinergic neurons in the A1 and A2 of the hindbrain. These findings may contribute to new therapeutic controls for hypothalamic ovulation disorders in humans and livestock.


Assuntos
Kisspeptinas , Receptores Purinérgicos P2 , Humanos , Ratos , Feminino , Animais , Kisspeptinas/metabolismo , Estradiol/farmacologia , Estradiol/metabolismo , Hormônio Luteinizante/metabolismo , Hipotálamo Anterior/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Estrogênios/farmacologia , Estrogênios/metabolismo , Neurônios/metabolismo , Ovulação , Rombencéfalo/metabolismo , Trifosfato de Adenosina/metabolismo , Nucleotídeos/metabolismo , Nucleotídeos/farmacologia , Adenosina/metabolismo
6.
Endocrinology ; 164(3)2023 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-36592113

RESUMO

Energy availability is an important regulator of reproductive function at various reproductive phases in mammals. Glucoprivation induced by 2-deoxy-D-glucose (2DG), an inhibitor of glucose utilization, as an experimental model of malnutrition suppresses the pulsatile release of GnRH/LH and induces gluconeogenesis. The present study was performed with the aim of examining whether enkephalin-δ-opioid receptor (DOR) signaling mediates the suppression of pulsatile GnRH/LH release and gluconeogenesis during malnutrition. The administration of naltrindole hydrochloride (NTI), a selective DOR antagonist, into the third ventricle blocked the suppression of LH pulses and part of gluconeogenesis induced by IV 2DG administration in ovariectomized rats treated with a negative feedback level of estradiol-17â€…ß (OVX + low E2). The IV 2DG administration significantly increased the number of Penk (enkephalin gene)-positive cells coexpressing fos (neuronal activation marker gene) in the paraventricular nucleus (PVN), but not in the arcuate nucleus (ARC) in OVX + low E2 rats. Furthermore, double in situ hybridization for Penk/Pdyn (dynorphin gene) in the PVN revealed that approximately 35% of the PVN Penk-expressing cells coexpressed Pdyn. Double in situ hybridization for Penk/Crh (corticotropin-releasing hormone gene) in the PVN and Penk/Kiss1 (kisspeptin gene) in the ARC revealed that few Penk-expressing cells coexpressed Crh and Kiss1. Taken together, these results suggest that central enkephalin-DOR signaling mediates the suppression of pulsatile LH release during malnutrition. Moreover, the current study suggests that central enkephalin-DOR signaling is also involved in gluconeogenesis during malnutrition in female rats.


Assuntos
Encefalinas , Gluconeogênese , Receptores Opioides delta , Animais , Feminino , Ratos , Núcleo Arqueado do Hipotálamo/metabolismo , Encefalinas/genética , Encefalinas/metabolismo , Glucose/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Hormônio Luteinizante/metabolismo , Hormônio Luteinizante/farmacologia , Mamíferos/metabolismo , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo
7.
Neurosci Lett ; 791: 136920, 2022 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-36272558

RESUMO

Follicular development and ovulation are profoundly suppressed during lactation. This suppression is suggested to be due to the suckling-induced inhibition of the kisspeptin gene (the master regulator of reproduction) in the arcuate nucleus (ARC) and subsequent inhibition of pulsatile gonadotropin-releasing hormone (GnRH)/gonadotropin release. The present study examined whether hypothalamic κ-opioid receptor (KOR) or µ-opioid receptor (MOR) signaling mediates the suppression of luteinizing hormone (LH) release induced by suckling stimulus during late lactation in rats. Central administration of a selective KOR antagonist blocked the suppression of LH release on Day 16 of lactation; however, central administration of a selective MOR antagonist failed to block the suppression. The suckling stimulus significantly increased the number of fos (a marker for neural activation)-positive Pdyn (dynorphin gene)-expressing cells in the paraventricular nucleus (PVN) and supraoptic nucleus (SON) but not in the ARC. Taken together, these results suggest that central KOR signaling, but not MOR signaling, at least partly, mediates the suppression of LH release induced by suckling stimulus during late lactation, and PVN and SON Dyn neurons may be involved in the suppression in rats.


Assuntos
Dinorfinas , Receptores Opioides kappa , Feminino , Ratos , Animais , Dinorfinas/metabolismo , Receptores Opioides kappa/metabolismo , Hormônio Luteinizante , Núcleo Arqueado do Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Lactação/fisiologia , Receptores Opioides
8.
Front Neurosci ; 16: 958377, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36033602

RESUMO

Endogenous opioid peptides have attracted attention as critical neuropeptides in the central mechanism regulating female reproduction ever since the discovery that arcuate dynorphin neurons that coexpress kisspeptin and neurokinin B (NKB), which are also known as kisspeptin/neurokinin B/dynorphin (KNDy) neurons, play a role as a master regulator of pulsatile gonadotropin-releasing hormone (GnRH) release in mammals. In this study, we first focus on the role of dynorphin released by KNDy neurons in the GnRH pulse generation. Second, we provide a historical overview of studies on endogenous opioid peptides. Third, we discuss how endogenous opioid peptides modulate tonic GnRH/gonadotropin release in female mammals as a mediator of inhibitory internal and external cues, such as ovarian steroids, nutritional status, or stress, on reproduction. Then, we discuss the role of endogenous opioid peptides in GnRH surge generation in female mammals.

9.
J Reprod Dev ; 68(3): 190-197, 2022 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-35249910

RESUMO

Reproductive function is suppressed during lactation owing to the suckling-induced suppression of the kisspeptin gene (Kiss1) expression in the arcuate nucleus (ARC) and subsequent suppression of luteinizing hormone (LH) release. Our previous study revealed that somatostatin (SST) neurons mediate suckling-induced suppression of LH release via SST receptor 2 (SSTR2) in ovariectomized lactating rats during early lactation. This study examined whether central SST-SSTR2 signaling mediates the inhibition of ARC Kiss1 expression and LH release in lactating rats during late lactation and whether the inhibition of glutamatergic neurons, stimulators of LH release, is involved in the suppression of LH release mediated by central SST-SSTR2 signaling in lactating rats. A central injection of the SSTR2 antagonist CYN154806 (CYN) significantly increased ARC Kiss1 expression in lactating rats on day 16 of lactation. Dual in situ hybridization revealed that few ARC Kiss1-positive cells co-expressed Sstr2, and some of the ARC Slc17a6 (a glutamatergic neuronal marker)-positive cells co-expressed Sstr2. Furthermore, almost all ARC Kiss1-positive cells co-expressed Grin1, a subunit of N-methyl-D-aspartate (NMDA) receptors. The numbers of Slc17a6/Sstr2 double-labeled and Slc17a6 single-labeled cells were significantly lower in lactating dams than in non-lactating rats whose pups had been removed after parturition. A central injection of an NMDA antagonist reversed the CYN-induced increase in LH release in lactating rats. Overall, these results suggest that central SST-SSTR2 signaling, at least partly, mediates the suppression of ARC Kiss1 expression and LH release by inhibiting ARC glutamatergic interneurons in lactating rats.


Assuntos
Interneurônios , Kisspeptinas , Lactação , Hormônio Luteinizante , Receptores de Somatostatina , Somatostatina , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Feminino , Interneurônios/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Lactação/metabolismo , Hormônio Luteinizante/metabolismo , N-Metilaspartato/metabolismo , Oligopeptídeos/farmacologia , Ratos , Receptores de Somatostatina/antagonistas & inibidores , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Somatostatina/genética , Somatostatina/metabolismo
10.
Diabetol Int ; 13(1): 35-40, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35059241

RESUMO

Suppression of hepatic gluconeogenesis is thought to largely underlie the antidiabetes action of metformin. However, this drug also exerts various effects on the gut, one of which is the enhancement of the uptake of 18F-labeled fluorodeoxyglucose (FDG), a nonmetabolizable glucose derivative, during [18F]FDG positron emission tomography (PET)-computed tomography (CT). Whereas the relevance of this effect to the glucose-lowering action of metformin remains unclear, it is of special interest because it was discovered in humans. Cessation of metformin treatment for several days is required to normalize [18F]FDG uptake in the intestine, suggesting that the enhanced uptake is not a direct effect of the drug in the circulation but rather a prolonged secondary effect. A recent study with state-of-the-art PET-magnetic resonance imaging (MRI), which provides better tissue registration and soft-tissue contrast compared with PET-CT, revealed that metformin-induced accumulation of [18F]FDG occurs primarily in the lumen of the intestine, indicating that the drug promotes excretion of glucose from the circulation into this space. This phenomenon does not necessarily imply that metformin stimulates the removal of glucose from the body in the stool. Instead, it might be related to changes in the abundance and metabolism of the gut microbiota induced by metformin. Further studies of this effect of metformin might shed light on the unanswered questions that still remain concerning the clinical action of this old drug.

11.
Int J Mol Sci ; 24(1)2022 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-36613635

RESUMO

Nuclear receptor subfamily 5 group A member 1 (NR5A1) is expressed in the pituitary gonadotrope and regulates their differentiation. Although several regulatory regions were implicated in Nr5a1 gene expression in the pituitary gland, none of these regions have been verified using mouse models. Furthermore, the molecular functions of NR5A1 in the pituitary gonadotrope have not been fully elucidated. In the present study, we generated mice lacking the pituitary enhancer located in the 6th intron of the Nr5a1 gene. These mice showed pituitary gland-specific disappearance of NR5A1, confirming the functional importance of the enhancer. Enhancer-deleted male mice demonstrated no defects at fetal stages. Meanwhile, androgen production decreased markedly in adult, and postnatal development of reproductive organs, such as the seminal vesicle, prostate, and penis was severely impaired. We further performed transcriptomic analyses of the whole pituitary gland of the enhancer-deleted mice and controls, as well as gonadotropes isolated from Ad4BP-BAC-EGFP mice. These analyses identified several genes showing gonadotrope-specific, NR5A1-dependent expressions, such as Spp1, Tgfbr3l, Grem1, and Nr0b2. These factors are thought to function downstream of NR5A1 and play important roles in reproductive organ development through regulation of pituitary gonadotrope functions.


Assuntos
Gonadotrofos , Hipófise , Sequências Reguladoras de Ácido Nucleico , Fator Esteroidogênico 1 , Animais , Masculino , Camundongos , Gonadotrofos/metabolismo , Íntrons/genética , Hipófise/metabolismo , Fator Esteroidogênico 1/genética
12.
Front Endocrinol (Lausanne) ; 12: 724632, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34566891

RESUMO

Increasing evidence accumulated during the past two decades has demonstrated that the then-novel kisspeptin, which was discovered in 2001, the known neuropeptides neurokinin B and dynorphin A, which were discovered in 1983 and 1979, respectively, and their G-protein-coupled receptors, serve as key molecules that control reproduction in mammals. The present review provides a brief historical background and a summary of our recent understanding of the roles of hypothalamic neurons expressing kisspeptin, neurokinin B, and dynorphin A, referred to as KNDy neurons, in the central mechanism underlying gonadotropin-releasing hormone (GnRH) pulse generation and subsequent tonic gonadotropin release that controls mammalian reproduction.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/fisiologia , Reprodução/fisiologia , Animais , Dinorfinas/metabolismo , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiologia , Kisspeptinas/metabolismo , Mamíferos , Neurocinina B/metabolismo , Neurônios/metabolismo
13.
Endocrinology ; 162(10)2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34270714

RESUMO

Energetic status often affects reproductive function, glucose homeostasis, and feeding in mammals. Malnutrition suppresses pulsatile release of the gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH) and increases gluconeogenesis and feeding. The present study aims to examine whether ß-endorphin-µ-opioid receptor (MOR) signaling mediates the suppression of pulsatile GnRH/LH release and an increase in gluconeogenesis/feeding induced by malnutrition. Ovariectomized female rats treated with a negative feedback level of estradiol-17ß (OVX + low E2) receiving 2-deoxy-D-glucose (2DG), an inhibitor of glucose utilization, intravenously (iv) were used as a malnutrition model. An administration of D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2 (CTOP), a selective MOR antagonist, into the third ventricle blocked the suppression of the LH pulse and increase in gluconeogenesis/feeding induced by iv 2DG administration. Histological analysis revealed that arcuate Kiss1 (kisspeptin gene)-expressing cells and preoptic Gnrh1 (GnRH gene)-expressing cells co-expressed little Oprm1 (MOR gene), while around 10% of arcuate Slc17a6 (glutamatergic marker gene)-expressing cells co-expressed Oprm1. Further, the CTOP treatment decreased the number of fos-positive cells in the paraventricular nucleus (PVN) in OVX + low E2 rats treated with iv 2DG but failed to affect the number of arcuate fos-expressing Slc17a6-positive cells. Taken together, these results suggest that the central ß-endorphin-MOR signaling mediates the suppression of pulsatile LH release and that the ß-endorphin may indirectly suppress the arcuate kisspeptin neurons, a master regulator for GnRH/LH pulses during malnutrition. Furthermore, the current study suggests that central ß-endorphin-MOR signaling is also involved in gluconeogenesis and an increase in food intake by directly or indirectly acting on the PVN neurons during malnutrition in female rats.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Luteinizante/metabolismo , Antagonistas de Entorpecentes/farmacologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores Opioides mu/metabolismo , beta-Endorfina/metabolismo , Animais , Glicemia/análise , Feminino , Gluconeogênese , Hipotálamo , Kisspeptinas/metabolismo , Ratos , Ratos Wistar , Receptores Opioides mu/biossíntese , Transdução de Sinais , Proteína Vesicular 2 de Transporte de Glutamato/biossíntese
14.
Peptides ; 142: 170546, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33794282

RESUMO

Metabolic stress resulting from either lack or excess of nutrients often causes infertility in both sexes. Kisspeptin-neurokinin B-dynorphin A (KNDy) neurons in the arcuate nucleus (ARC) has been suggested to be a key players in reproduction via direct stimulation of the pulsatile gonadotropin-releasing hormone (GnRH) and subsequent gonadotropin release in mammalian species. In this study, we investigated the effect of high-fat diet (HFD) on hypothalamic KNDy gene expression to examine the pathogenic mechanism underlying obesity-induced infertility in male and female rats. Male and female rats at 7 weeks of age were fed with either a standard or HFD for 4 months. In the male rats, the HFD caused a significant suppression of ARC Kiss1 and Pdyn gene expressions, but did not affect the plasma luteinizing hormone (LH) levels and sizes of the morphology of the testis and epididymis. In the female rats, 58% of the HFD-fed female rats exhibited irregular estrous cycles, whereas the remaining rats showed regular cycles. Two of the 10 rats that showed HFD-induced irregular estrous cycles showed profound suppression of LH pulse frequency and the number of ARC Kiss1-expressing cells, whereas the other females showed normal LH pulses and ARC Kiss1 expression. Our finding shows that suppression of ARC Kiss1 expression might be the initial pathological change of hypogonadotropic hypogonadism in HFD-fed male rats, while the obese-related infertility in the female rats may be mainly induced by KNDy-independent pathways. Taken together, ARC kisspeptin neurons in male rats may be susceptible to HFD-induced obesity compared with those in female rats.


Assuntos
Núcleo Arqueado do Hipotálamo/patologia , Hormônios Esteroides Gonadais/farmacologia , Hipogonadismo/patologia , Hormônio Luteinizante/metabolismo , Doenças Metabólicas/complicações , Neurônios/patologia , Obesidade/complicações , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Dieta Hiperlipídica , Dinorfinas/genética , Dinorfinas/metabolismo , Feminino , Hipogonadismo/etiologia , Hipogonadismo/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Masculino , Neurocinina B/genética , Neurocinina B/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Ratos Wistar
15.
Proc Natl Acad Sci U S A ; 118(5)2021 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-33500349

RESUMO

The gonadotropin-releasing hormone (GnRH) pulse is fundamental for mammalian reproduction: GnRH pulse regimens are needed as therapies for infertile women as continuous GnRH treatment paradoxically inhibits gonadotropin release. Circumstantial evidence suggests that the hypothalamic arcuate KNDy neurons expressing kisspeptin (encoded by Kiss1), neurokinin B (encoded by Tac3), and dynorphin A serve as a GnRH pulse generator; however, no direct evidence is currently available. Here, we show that rescuing >20% KNDy neurons by transfecting Kiss1 inside arcuate Tac3 neurons, but not outside of these neurons, recovered folliculogenesis and luteinizing hormone (LH) pulses, an indicator of GnRH pulses, in female global Kiss1 knockout (KO) rats and that >90% conditional arcuate Kiss1 KO in newly generated Kiss1-floxed rats completely suppressed LH pulses. These results first provide direct evidence that KNDy neurons are the GnRH pulse generator, and at least 20% of KNDy neurons are sufficient to maintain folliculogenesis via generating GnRH/gonadotropin pulses.


Assuntos
Dinorfinas/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Gonadotropinas/metabolismo , Kisspeptinas/metabolismo , Neurocinina B/metabolismo , Neurônios/metabolismo , Organogênese , Folículo Ovariano/crescimento & desenvolvimento , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Aromatase/genética , Aromatase/metabolismo , Retroalimentação Fisiológica , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Integrases/metabolismo , Hormônio Luteinizante/sangue , Tamanho do Órgão , Folículo Ovariano/metabolismo , Hipófise/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Receptores do LH/genética , Receptores do LH/metabolismo , Receptores LHRH/metabolismo
16.
Cancer Sci ; 111(12): 4359-4370, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32976661

RESUMO

Metastatic progression remains the major cause of death in human breast cancer. Cancer cells with cancer stem cell (CSC) properties drive initiation and growth of metastases at distant sites. We have previously established the breast cancer patient-derived tumor xenograft (PDX) mouse model in which CSC marker CD44+ cancer cells formed spontaneous microscopic metastases in the liver. In this PDX mouse, the expression levels of S100A10 and its family proteins were much higher in the CD44+ cancer cells metastasized to the liver than those at the primary site. Knockdown of S100A10 in breast cancer cells suppressed and overexpression of S100A10 in breast cancer PDX cells enhanced their invasion abilities and 3D organoid formation capacities in vitro. Mechanistically, S100A10 regulated the matrix metalloproteinase activity and the expression levels of stem cell-related genes. Finally, constitutive knockdown of S100A10 significantly reduced their metastatic ability to the liver in vivo. These findings suggest that S100A10 functions as a metastasis promoter of breast CSCs by conferring both invasion ability and CSC properties in breast cancers.


Assuntos
Anexina A2/metabolismo , Neoplasias da Mama/metabolismo , Células-Tronco Neoplásicas/metabolismo , Proteínas S100/metabolismo , Regulação para Cima , Animais , Anexina A2/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Perfilação da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Receptores de Hialuronatos/metabolismo , Lentivirus/genética , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/secundário , Metaloproteinases da Matriz/metabolismo , Camundongos , Invasividade Neoplásica , Metástase Neoplásica , Células-Tronco Neoplásicas/patologia , Organoides , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas S100/genética
17.
Endocrinology ; 161(11)2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32894768

RESUMO

Malnutrition suppresses reproductive functions in mammals, which is considered to be mostly due to the inhibition of pulsatile gonadotropin-releasing hormone (GnRH)/gonadotropin secretion. Accumulating evidence suggests that kisspeptin neurons in the arcuate nucleus (ARC) play a critical role in the regulation of pulsatile GnRH/gonadotropin release. The present study aimed to examine if the hypothalamic dynorphin A (Dyn) neurons mediate the suppression of GnRH/luteinizing hormone (LH) pulses during malnutrition. Ovariectomized rats treated with a negative feedback level of estradiol-17ß-treated (OVX+E2) were administered with intravenous (iv) or fourth cerebroventricle (4V) 2-deoxy-D-glucose (2DG), an inhibitor of glucose utilization, to serve as a malnutrition model. Central administration of a Dyn receptor antagonist blocked the iv- or 4V-2DG-induced suppression of LH pulses in OVX+E2 rats. The 4V 2DG administration significantly increased the number of Pdyn (Dyn gene)-positive cells co-expressing fos in the paraventricular nucleus (PVN), but not in the ARC and supraoptic nucleus (SON), and the iv 2DG treatment significantly increased the number of fos and Pdyn-co-expressing cells in the PVN and SON, but decreased it in the ARC. The E2 treatment significantly increased Pdyn expression in the PVN, but not in the ARC and SON. Double in situ hybridization for Kiss1 (kisspeptin gene) and Oprk1 (Dyn receptor gene) revealed that around 60% of ARC Kiss1-expressing cells co-expressed Oprk1. These results suggest that the PVN Dyn neurons, at least in part, mediate LH pulse suppression induced by the hindbrain or peripheral glucoprivation, and Dyn neurons may directly suppress the ARC kisspeptin neurons in female rats.


Assuntos
Dinorfinas/metabolismo , Privação de Alimentos/fisiologia , Hormônio Luteinizante/metabolismo , Neurônios/fisiologia , Núcleo Hipotalâmico Paraventricular , Rombencéfalo/metabolismo , Animais , Regulação para Baixo/efeitos dos fármacos , Feminino , Glucose/metabolismo , Glucose/farmacologia , Kisspeptinas/metabolismo , Desnutrição/metabolismo , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos , Ratos Wistar , Rombencéfalo/efeitos dos fármacos
18.
Endocrinology ; 160(2): 473-483, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30544226

RESUMO

Follicular development and ovulation are profoundly suppressed during lactation in mammals. This suppression is suggested to be mainly due to the suckling-induced inhibition of kisspeptin gene (Kiss1) expression in the arcuate nucleus (ARC) and consequent inhibition of pulsatile GnRH/LH release. We examined whether central somatostatin (SST) signaling mediates the suckling-induced suppression of pulsatile LH secretion. SST has been reported to be expressed in the posterior intralaminar thalamic nucleus (PIL), where the suckling stimulus is postulated to be relayed to the hypothalamus during lactation. SST inhibitory receptors (SSTRs) are abundantly expressed in the ARC, where kisspeptin/neurokinin B/dynorphin A (KNDy) neurons are located. Histological and quantitative studies revealed that the suckling stimulus increased the number of SST-expressing cells in the PIL, and Sstr2 expression in the ARC. Furthermore, a central injection of an SSTR2 antagonist caused a significant increase in pulsatile LH release in lactating rats. Double labeling of Sstr2 and the neurokinin B gene, as a marker for ARC KNDy neurons, showed Sstr2 expression was abundantly detected in the ARC, but few KNDy neurons coexpressed Sstr2 in lactating rats. Taken together, these findings suggest the suckling-induced activation of SST-SSTR2 signaling mediates, at least in part, the suppression of pulsatile LH secretion during lactation in rats, probably via the indirect effects of SST on KNDy neurons. These results provide a new aspect on the role of central SST-SSTR signaling in understanding the mechanism underlying lactational anestrus.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Lactação , Hormônio Luteinizante/metabolismo , Receptores de Somatostatina/metabolismo , Animais , Animais Lactentes , Feminino , Núcleos Intralaminares do Tálamo/metabolismo , Neuropeptídeos/metabolismo , Ratos , Receptores de Somatostatina/antagonistas & inibidores , Somatostatina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...