Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.120
Filtrar
1.
Endocrinology ; 165(10)2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39253941

RESUMO

Reproductive function in mammals depends on the ability of progesterone (P4) to suppress pulsatile gonadotrophin-releasing hormone (GnRH) and luteinizing hormone (LH) secretion in a homeostatic-negative feedback loop. Previous research identified that cells upstream from GnRH neurons expressing the nuclear progesterone receptor (PGR) are required for P4-negative feedback. However, the identity of these cells and the mechanism by which they reduce GnRH/LH pulsatile secretion is unknown. We aimed to address the hypothesis that PGR expressed by a neural population in the arcuate nucleus recently identified as the GnRH pulse generator, cells expressing kisspeptin, neurokinin B, and dynorphin (KNDy cells), mediate P4-negative feedback. To achieve this, we used female mice with the PGR gene conditionally deleted from kisspeptin cells (KPRKO mice) and observed a substantial decrease in the percentage of KNDy neurons coexpressing PGR messenger RNA (mRNA) (11% in KPRKO mice vs 86% in wild-type [WT] mice). However, KPRKO mice did not display changes in the frequency or amplitude of LH pulses in diestrus or estrus, nor in the ability of exogenous P4 to blunt a postcastration increase in LH. Further, mRNA expression of arcuate kisspeptin and dynorphin, which are excitatory and inhibitory to GnRH secretion, respectively, remained unaltered in KPRKO mice compared to WT controls. Together, these findings show that the near-complete loss of PGR signaling from KNDy cells does not affect negative feedback regulation of GnRH pulse generation in mice, suggesting that feedback through this receptor can occur via a small number of KNDy cells or a yet unidentified cell population.


Assuntos
Núcleo Arqueado do Hipotálamo , Retroalimentação Fisiológica , Hormônio Liberador de Gonadotropina , Kisspeptinas , Hormônio Luteinizante , Camundongos Knockout , Progesterona , Receptores de Progesterona , Animais , Feminino , Kisspeptinas/metabolismo , Kisspeptinas/genética , Receptores de Progesterona/metabolismo , Receptores de Progesterona/genética , Hormônio Luteinizante/metabolismo , Camundongos , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Liberador de Gonadotropina/genética , Núcleo Arqueado do Hipotálamo/metabolismo , Progesterona/metabolismo , Dinorfinas/metabolismo , Dinorfinas/genética , Neurônios/metabolismo , Neurocinina B/genética , Neurocinina B/metabolismo
2.
Endocrinology ; 165(10)2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39254333

RESUMO

There has been an alarming trend toward earlier puberty in girls, suggesting the influence of an environmental factor(s). As the reactivation of the reproductive axis during puberty is thought to be mediated by the hypothalamic neuropeptides kisspeptin and gonadotropin-releasing hormone (GnRH), we asked whether an environmental compound might activate the kisspeptin (KISS1R) or GnRH receptor (GnRHR). We used GnRHR or KISS1R-expressing HEK293 cells to screen the Tox21 10K compound library, a compendium of pharmaceuticals and environmental compounds, for GnRHR and KISS1R activation. Agonists were identified using Ca2+ flux and phosphorylated extracellularly regulated kinase (p-ERK) detection assays. Follow-up studies included measurement of genes known to be upregulated upon receptor activation using relevant murine or human cell lines and molecular docking simulation. Musk ambrette was identified as a KISS1R agonist, and treatment with musk ambrette led to increased expression of Gnrh1 in murine and human hypothalamic cells and expansion of GnRH neuronal area in developing zebrafish larvae. Molecular docking demonstrated that musk ambrette interacts with the His309, Gln122, and Gln123 residues of the KISS1R. A group of cholinergic agonists with structures similar to methacholine was identified as GnRHR agonists. When applied to murine gonadotrope cells, these agonists upregulated Fos, Jun, and/or Egr1. Molecular docking revealed a potential interaction between GnRHR and 5 agonists, with Asn305 constituting the most conservative GnRHR binding site. In summary, using a Tox21 10K compound library screen combined with cellular, molecular, and structural biology techniques, we have identified novel environmental agents that may activate the human KISS1R or GnRHR.


Assuntos
Receptores de Kisspeptina-1 , Receptores LHRH , Humanos , Feminino , Animais , Receptores de Kisspeptina-1/metabolismo , Receptores de Kisspeptina-1/genética , Receptores LHRH/metabolismo , Receptores LHRH/genética , Camundongos , Células HEK293 , Peixe-Zebra , Hormônio Liberador de Gonadotropina/metabolismo , Puberdade/efeitos dos fármacos , Hipotálamo/metabolismo , Hipotálamo/efeitos dos fármacos , Simulação de Acoplamento Molecular , Maturidade Sexual/efeitos dos fármacos , Maturidade Sexual/fisiologia , Kisspeptinas/metabolismo , Kisspeptinas/genética , Poluentes Ambientais/toxicidade , Poluentes Ambientais/farmacologia
3.
Ceska Gynekol ; 89(4): 269-277, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39242201

RESUMO

OBJECTIVE: Endometrial polyp (EP) is a type of pathology that is quite common in clinical practice. Although its exact etiology is not fully known, there is evidence to support that it is sensitive to hormonal stimuli. We aimed to investigate the relationship between kisspeptin (KP) and EP by comparing the genetic (tissue-blood) and immunohistochemical (IHC) expression of KP in EP lesions in patients with normal endometrial findings. MATERIALS AND METHODS: A prospective case-control study of 50 patients with EP (N = 25) and normal endometrial findings (N = 25) on biopsy and/or excision material was performed. Blood and biopsy samples obtained from all patients were stored at -80 °C. KP gene expression levels were determined from paraffin blocks, and peripheral venous blood samples obtained from biopsy specimens and IHC-H-score analysis were performed from paraffin blocks. EP and matched controls were compared for KP. RESULTS: After IHC, the KP H-score of the control group was higher than the EP group, and this difference was statistically significant; H-score: control: 5 (++; 1-15); polyp: 1 (+; 0-12) (P < 0.05). Although KP expression in both tissue and blood was higher in the control group than in the EP group, this difference was not statistically significant (P > 0.05). No significant correlation was found between IHC H-score and KP expression levels in tissue and blood. According to the ROC analysis, the tissue and blood KP expression cut-off value and area under the curve (AUC) predicting the likelihood of developing EP were not significant (tissue KP: 1.04, AUC: 0.570, P = 0.388, sensitivity 56%, specificity 60%, Blood KP: 1.06, AUC: 0.569, P = 0.401, sensitivity 80%, specificity 40%). CONCLUSIONS: Decreased KP expression level in EP lesions may predict the diagnosis of EP, and in the future, KP may have therapeutic potential for benign gynecological pathologies such as polyps.


Assuntos
Imuno-Histoquímica , Kisspeptinas , Pólipos , Humanos , Feminino , Pólipos/genética , Pólipos/metabolismo , Pólipos/patologia , Kisspeptinas/genética , Kisspeptinas/metabolismo , Estudos de Casos e Controles , Doenças Uterinas/genética , Doenças Uterinas/metabolismo , Doenças Uterinas/patologia , Doenças Uterinas/sangue , Estudos Prospectivos , Adulto , Endométrio/metabolismo , Endométrio/patologia , Pessoa de Meia-Idade
4.
Phytomedicine ; 133: 155931, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39116604

RESUMO

BACKGROUND: Polycystic ovary syndrome (PCOS) is one of the most common reproductive endocrine disorders. Accumulated evidence has suggested the indispensable role of kisspeptin-G protein-coupled receptor (GPR54) system and SHBG in development of PCOS. However, potential mechanisms and their relationship are unclear. Jiawei Buzhong Yiqi Decoction (JWBZYQ) has been reported to ameliorate obese PCOS. Whereas, potential mechanisms remain elusive. PURPOSE: To determine whether JWBZYQ attenuates PCOS by regulating the kisspeptin-GPR54 system and SHBG production. And to explore potential mechanisms. METHODS: An overweight PCOS rat model was developed with testosterone propionate (TP) and high-fat diet (HFD). The efficacy of JWBZYQ was assessed by tracking changes in weight, estrous cycle, ovarian morphology, and serum sex hormone levels. Additionally, kisspeptin-GPR54 system expression in multiple organs and PI3K-AKT pathway activity in liver of different rats were detected. Modifications in SHBG production were also measured. Kisspeptin54 was administered to establish a cellular model. The levels of AKT phosphorylation and SHBG protein within HepG2 cells were analyzed. Finally, confirmatory studies were performed using AKT phosphorylation activator and inhibitor. RESULTS: JWBZYQ effectively attenuated the overweight, disrupted estrous cycle, altered sex hormone levels, and aberrant ovarian morphology in PCOS rats. Meanwhile, PCOS rats exhibited elevated levels of kisspeptin and GPR54, along with reduced SHBG levels, which could be reversed by JWBZYQ. These alterations might be connected with the activation of AKT phosphorylation. In vitro experiment identified that JWBZYQ could rectify the hyperactivated AKT phosphorylation and deficient production of SHBG caused by kisspeptin54. CONCLUSIONS: Overexpressed kisspeptin-GPR54 system inhibited SHBG synthesis in PCOS. JWBZYQ curtailed the exorbitant expression of kisspeptin and GPR54, which moderated the rise in AKT phosphorylation and subsequently promoted the production of SHBG.


Assuntos
Medicamentos de Ervas Chinesas , Kisspeptinas , Síndrome do Ovário Policístico , Proteínas Proto-Oncogênicas c-akt , Ratos Sprague-Dawley , Receptores de Kisspeptina-1 , Globulina de Ligação a Hormônio Sexual , Síndrome do Ovário Policístico/tratamento farmacológico , Síndrome do Ovário Policístico/metabolismo , Animais , Feminino , Kisspeptinas/metabolismo , Medicamentos de Ervas Chinesas/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Kisspeptina-1/metabolismo , Globulina de Ligação a Hormônio Sexual/metabolismo , Ratos , Modelos Animais de Doenças , Dieta Hiperlipídica , Ovário/efeitos dos fármacos , Ovário/metabolismo , Transdução de Sinais/efeitos dos fármacos , Humanos , Receptores Acoplados a Proteínas G/metabolismo , Propionato de Testosterona
5.
Artigo em Inglês | MEDLINE | ID: mdl-39089445

RESUMO

Temperature is a preeminent factor in the regulation of fish reproduction and hinders gonadal development beyond a specific threshold. To comprehend the molecular mechanism responsible for reproductive suppression at different temperature, expression of the genes encoding kisspeptin (kiss2), gonadotropin-releasing hormone (gnrh1) and their receptors (gpr54, gnrh1r) in the brain, and the gonadotropin (GTH) subunits (fshb and lhb) in the pituitary were studied in juvenile Nile tilapia (Oreochromis niloticus) along with gonadal histology. Fish were acclimatized to three distinct temperatures, including 31 °C, 34 °C and 37 °C for 14 days. The mRNA levels of kiss2, gpr54, gnrh1, and gnrh1r were significantly decreased at 37 °C compared to 31 °C and 34 °C in the both sexes. In parallel, the expression level of fshb in the both sexes and lhb in the female were significantly lower at 37 °C in the pituitary. Histologically, the gonads of both sexes had normal growth of gametes at control temperature (31 °C), whereas the spermatogenesis and oocyte maturation were slowed down and atretic oocytes were found in the ovary at 37 °C acclimation temperature. Taken together, the results imply that elevated temperature beyond the specific threshold may have a negative impact on reproduction by suppressing the gene expressions of kisspeptin/GnRH1/GTH system and eventually restrains normal growth and maturation of gametes in the both sexes of Nile tilapia.


Assuntos
Ciclídeos , Hormônio Liberador de Gonadotropina , Gônadas , Kisspeptinas , Animais , Kisspeptinas/genética , Kisspeptinas/metabolismo , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Ciclídeos/genética , Ciclídeos/crescimento & desenvolvimento , Ciclídeos/metabolismo , Feminino , Masculino , Gônadas/metabolismo , Gônadas/crescimento & desenvolvimento , Temperatura , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Hipófise/metabolismo , Ovário/metabolismo , Ovário/crescimento & desenvolvimento , Gonadotropinas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento
6.
Biomolecules ; 14(8)2024 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-39199311

RESUMO

Kisspeptin, a key neuropeptide derived from the KISS1R gene, is renowned for its critical role in regulating the hypothalamic-pituitary-gonadal axis and reproductive hormone secretion. Beyond its primary function in reproductive biology, emerging research has illuminated its influence in various cancers, mediating significant effects through its interaction with the G protein-coupled receptor, kisspeptin receptor. This interaction has been implicated in modulating cellular processes such as proliferation and metastasis, making it a potential target for therapeutic intervention. Our study initially screened ten kisspeptin-10 analogs through cytotoxic effects of kisspeptin-10 (KP10) and its analogs in several cancer types, including cervical, prostate, breast, and gastric cancers, with a particular focus on cervical cancer, where the most profound effects were observed. Further exploration using kinase array assays revealed that these analogs specifically alter key kinases involved in cancer progression. Migration assays demonstrated a substantial decrease in cell motility, and Bioluminescence Resonance Energy Transfer assays confirmed these analogs' strong interactions with the kisspeptin receptor. Overall, our results indicate that these KP10 analogs not only hinder cervical cancer cell proliferation but also curtail migration through targeted modulation of kinase signaling, suggesting their potential as therapeutic agents in managing cervical cancer progression. This comprehensive approach underscores the therapeutic promise of exploiting kisspeptin signaling in cancer treatment strategies.


Assuntos
Kisspeptinas , Transdução de Sinais , Neoplasias do Colo do Útero , Kisspeptinas/metabolismo , Kisspeptinas/genética , Kisspeptinas/farmacologia , Humanos , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/patologia , Neoplasias do Colo do Útero/tratamento farmacológico , Neoplasias do Colo do Útero/genética , Feminino , Transdução de Sinais/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Receptores de Kisspeptina-1/metabolismo , Receptores de Kisspeptina-1/genética
7.
Sci Adv ; 10(33): eadn7771, 2024 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-39151001

RESUMO

Kisspeptin receptor (KISS1R), belonging to the class A peptide-GPCR family, plays a key role in the regulation of reproductive physiology after stimulation by kisspeptin and is regarded as an attractive drug target for reproductive diseases. Here, we demonstrated that KISS1R can couple to the Gi/o pathway besides the well-known Gq/11 pathway. We further resolved the cryo-electron microscopy (cryo-EM) structure of KISS1R-Gq and KISS1R-Gi complexes bound to the synthetic agonist TAK448 and structure of KISS1R-Gq complex bound to the endogenous agonist KP54. The high-resolution structures provided clear insights into mechanism of KISS1R recognition by its ligand and can facilitate the design of targeted drugs with high affinity to improve treatment effects. Moreover, the structural and functional analyses indicated that conformational differences in the extracellular loops (ECLs), intracellular loops (ICLs) of the receptor, and the "wavy hook" of the Gα subunit may account for the specificity of G protein coupling for KISS1R signaling.


Assuntos
Microscopia Crioeletrônica , Receptores de Kisspeptina-1 , Humanos , Ligantes , Receptores de Kisspeptina-1/metabolismo , Receptores de Kisspeptina-1/química , Ligação Proteica , Kisspeptinas/metabolismo , Kisspeptinas/química , Modelos Moleculares , Células HEK293 , Conformação Proteica , Transdução de Sinais , Proteínas de Ligação ao GTP/metabolismo , Proteínas de Ligação ao GTP/química , Relação Estrutura-Atividade
8.
Neurosci Lett ; 837: 137918, 2024 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-39096756

RESUMO

Neurons co-expressing kisspeptin, neurokinin B, and dynorphin A (KNDy neurons), located in the arcuate nucleus (ARC) of the hypothalamus, are indicated to be the gonadotropin-releasing hormone (GnRH) pulse generator. Dynorphin A is reported to suppress GnRH pulse generator activity. Nalfurafine is a selective agonist of the κ-opioid receptor (KOR), a receptor for dynorphin A, clinically used as an anti-pruritic drug. This study aimed to evaluate the effects of nalfurafine on GnRH pulse generator activity and luteinizing hormone (LH) pulses using female goats. Nalfurafine (0, 2, 4, 8, or 16 µg/head) was intravenously injected into ovariectomized Shiba goats. The multiple unit activity (MUA) in the ARC area was recorded, and plasma LH concentrations were measured 2 and 48 h before and after injection, respectively. The MUA volley interval during 0-2 h after injection was significantly increased in the nalfurafine 8 and 16 µg groups compared with the vehicle group. In 0-2 h after injection, the number of LH pulses was significantly decreased in the nalfurafine 8 and 16 µg groups, and the mean and baseline LH were significantly decreased in all nalfurafine-treated groups (2, 4, 8, and 16 µg) compared with the vehicle group. These results suggest that nalfurafine inhibits the activity of the GnRH pulse generator in the ARC, thus suppressing pulsatile LH secretion. Therefore, nalfurafine could be used as a reproductive inhibitor in mammals.


Assuntos
Núcleo Arqueado do Hipotálamo , Cabras , Hormônio Liberador de Gonadotropina , Morfinanos , Receptores Opioides kappa , Compostos de Espiro , Animais , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/metabolismo , Feminino , Compostos de Espiro/farmacologia , Compostos de Espiro/administração & dosagem , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Liberador de Gonadotropina/agonistas , Morfinanos/farmacologia , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Hormônio Luteinizante/sangue , Hormônio Luteinizante/metabolismo , Kisspeptinas/metabolismo , Dinorfinas/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurocinina B/metabolismo
9.
J Clin Invest ; 134(15)2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-39087475

RESUMO

Kisspeptin is an essential neuropeptide sitting at the apex of the hypothalamo-pituitary-gonadal (HPG) endocrine axis to regulate gonadotropin-releasing hormone (GnRH) neurons and downstream reproductive hormones. Kisspeptin neurons integrate feedback from sex steroids facilitating regulation of the menstrual cycle and mediate the effects of metabolic stressors on the reproductive axis. In this issue of the JCI, Torres and colleagues describe another pathway for kisspeptin signaling in astrocytes to influence GnRH neuronal output. Astrocytes had kisspeptin receptors that activated canonical intracellular signaling pathways to constrain the magnitude of kisspeptin-induced GnRH neuronal stimulation. Additionally, the appositions between kisspeptin and GnRH neurons were dynamic during the ovarian cycle, with astrocyte kisspeptin signaling proposed as a putative modulator of this neuroplasticity. Importantly, astrocyte kisspeptin signaling also mediated susceptibility to metabolic stressors and the development of obesity-induced hypogonadism, underscoring the physiological and pathological importance of this pathway and revealing the importance of nonneuronal signaling in reproductive health.


Assuntos
Astrócitos , Hormônio Liberador de Gonadotropina , Kisspeptinas , Neurônios , Transdução de Sinais , Astrócitos/metabolismo , Humanos , Hormônio Liberador de Gonadotropina/metabolismo , Animais , Feminino , Neurônios/metabolismo , Kisspeptinas/metabolismo , Ciclo Menstrual/metabolismo , Reprodução
10.
Hum Reprod ; 39(9): 2089-2103, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38978296

RESUMO

STUDY QUESTION: Do hyperactive kisspeptin neurons contribute to abnormally high LH secretion and downstream hyperandrogenemia in polycystic ovary syndrome (PCOS)-like conditions and can inhibition of kisspeptin neurons rescue such endocrine impairments? SUMMARY ANSWER: Targeted inhibition of endogenous kisspeptin neuron activity in a mouse model of PCOS reduced the abnormally hyperactive LH pulse secretion and hyperandrogenemia to healthy control levels. WHAT IS KNOWN ALREADY: PCOS is a reproductive disorder characterized by hyperandrogenemia, anovulation, and/or polycystic ovaries, along with a hallmark feature of abnormal LH hyper-pulsatility, but the mechanisms underlying the endocrine impairments remain unclear. A chronic letrozole (LET; aromatase inhibitor) mouse model recapitulates PCOS phenotypes, including polycystic ovaries, anovulation, high testosterone, and hyperactive LH pulses. LET PCOS-like females also have increased hypothalamic kisspeptin neuronal activation which may drive their hyperactive LH secretion and hyperandrogenemia, but this has not been tested. STUDY DESIGN, SIZE, DURATION: Transgenic KissCRE+/hM4Di female mice or littermates Cre- controls were treated with placebo, or chronic LET (50 µg/day) to induce a PCOS-like phenotype, followed by acute (once) or chronic (2 weeks) clozapine-N-oxide (CNO) exposure to chemogenetically inhibit kisspeptin cells (n = 6 to 10 mice/group). PARTICIPANTS/MATERIALS, SETTING, METHODS: Key endocrine measures, including in vivo LH pulse secretion patterns and circulating testosterone levels, were assessed before and after selective kisspeptin neuron inhibition and compared between PCOS groups and healthy controls. Alterations in body weights were measured and pituitary and ovarian gene expression was determined by qRT-PCR. MAIN RESULTS AND THE ROLE OF CHANCE: Acute targeted inhibition of kisspeptin neurons in PCOS mice successfully lowered the abnormally hyperactive LH pulse secretion (P < 0.05). Likewise, chronic selective suppression of kisspeptin neuron activity reversed the previously high LH and testosterone levels (P < 0.05) down to healthy control levels and rescued reproductive gene expression (P < 0. 05). LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: Ovarian morphology was not assessed in this study. Additionally, mouse models can offer mechanistic insights into neuroendocrine processes in PCOS-like conditions but may not perfectly mirror PCOS in women. WIDER IMPLICATIONS OF THE FINDINGS: These data support the hypothesis that overactive kisspeptin neurons can drive neuroendocrine PCOS-like impairments, and this may occur in PCOS women. Our findings complement recent clinical investigations using NKB receptor antagonists to lower LH in PCOS women and suggest that pharmacological dose-dependent modulation of kisspeptin neuron activity may be a valuable future therapeutic target to clinically treat hyperandrogenism and lower elevated LH in PCOS women. STUDY FUNDING/COMPETING INTEREST(S): This research was supported by NIH grants R01 HD111650, R01 HD090161, R01 HD100580, P50 HD012303, R01 AG078185, and NIH R24 HD102061, and a pilot project award from the British Society for Neuroendocrinology. There are no competing interests.


Assuntos
Modelos Animais de Doenças , Hiperandrogenismo , Kisspeptinas , Letrozol , Hormônio Luteinizante , Neurônios , Síndrome do Ovário Policístico , Animais , Síndrome do Ovário Policístico/metabolismo , Feminino , Kisspeptinas/metabolismo , Hormônio Luteinizante/sangue , Hiperandrogenismo/metabolismo , Hiperandrogenismo/complicações , Camundongos , Neurônios/metabolismo , Letrozol/farmacologia , Camundongos Transgênicos , Inibidores da Aromatase/farmacologia , Testosterona/sangue
11.
J Neurosci ; 44(34)2024 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-39038954

RESUMO

Stress impairs fertility, at least in part, via inhibition of gonadotropin secretion. Luteinizing hormone (LH) is an important gonadotropin that is released in a pulsatile pattern in males and in females throughout the majority of the ovarian cycle. Several models of stress, including acute metabolic stress, suppress LH pulses via inhibition of neurons in the arcuate nucleus of the hypothalamus that coexpress kisspeptin, neurokinin B, and dynorphin (termed KNDy cells) which form the pulse generator. The mechanism for inhibition of KNDy neurons during stress, however, remains a significant outstanding question. Here, we investigated a population of catecholamine neurons in the nucleus of the solitary tract (NTS), marked by expression of the enzyme dopamine beta-hydroxylase (DBH), in female mice. First, we found that a subpopulation of DBH neurons in the NTS is activated (express c-Fos) during metabolic stress. Then, using chemogenetics, we determined that activation of these cells is sufficient to suppress LH pulses, augment corticosterone secretion, and induce sickness-like behavior. In subsequent studies, we identified evidence for suppression of KNDy cells (rather than downstream signaling pathways) and determined that the suppression of LH pulses was not dependent on the acute rise in glucocorticoids. Together these data support the hypothesis that DBH cells in the NTS are important for regulation of neuroendocrine and behavioral responses to stress.


Assuntos
Hormônio Luteinizante , Núcleo Solitário , Animais , Feminino , Hormônio Luteinizante/metabolismo , Camundongos , Núcleo Solitário/metabolismo , Dopamina beta-Hidroxilase/metabolismo , Camundongos Endogâmicos C57BL , Neurônios Adrenérgicos/metabolismo , Neurônios Adrenérgicos/fisiologia , Corticosterona/metabolismo , Norepinefrina/metabolismo , Camundongos Transgênicos , Estresse Fisiológico/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Kisspeptinas/metabolismo , Neurocinina B/metabolismo
12.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(3): 542-551, 2024 May 20.
Artigo em Chinês | MEDLINE | ID: mdl-38948287

RESUMO

Objective: Kisspeptin, a protein encoded by the KISS1 gene, functions as an essential factor in suppressing tumor growth. The intricate orchestration of cellular processes such as proliferation and differentiation is governed by the Notch1/Akt/Foxo1 signaling pathway, which assumes a central role in maintaining cellular homeostasis. In the specific context of this investigation, the focal point lies in a meticulous exploration of the intricate mechanisms underlying the regulatory effect of kisspeptin on the process of endometrial decidualization. This investigation delves into the interplay between kisspeptin and the Notch1/Akt/Foxo1 signaling pathway, aiming to elucidate its significance in the pathophysiology of recurrent spontaneous abortion (RSA). Methods: We enrolled a cohort comprising 45 individuals diagnosed with RSA, who were admitted to the outpatient clinic of the Reproductive Center at the Second Affiliated Hospital of Soochow University between June 2020 and December 2020. On the other hand, an additional group of 50 women undergoing elective abortion at the outpatient clinic of the Family Planning Department during the same timeframe was also included. To comprehensively assess the molecular landscape, Western blot and RT-qPCR were performed to analyze the expression levels of kisspeptin (and its gene KISS1), IGFBP1 (an established marker of decidualization), Notch1, Akt, and Foxo1 within the decidua. Human endometrial stromal cells (hESC) were given targeted interventions, including treatment with siRNA to disrupt KISS1 or exposure to kisspeptin10 (the bioactive fragment of kisspeptin), and were subsequently designated as the siKP group or the KP10 group, respectively. A control group comprised hESC was transfected with blank siRNA, and cell proliferation was meticulously evaluated with CCK8 assay. Following in vitro induction for decidualization across the three experimental groups, immunofluorescence assay was performed to identify differences in Notch1 expression and decidualization morphology between the siKP and the KP10 groups. Furthermore, RT-qPCR and Western blot were performed to gauge the expression levels of IGFBP1, Notch1, Akt, and Foxo1 across the three cell groups. Subsequently, decidualization was induced in hESC by adding inhibitors targeting Notch1, Akt, and Foxo1. The expression profiles of the aforementioned proteins and genes in the four groups were then examined, with hESC induced for decidualization without adding inhibitors serving as the normal control group. To establish murine models of normal pregnancy (NP) and RSA, CBA/J×BALB/c and CBA/J×DBA/2 mice were used. The mice were respectively labeled as the NP model and RSA model. The experimental groups received intraperitoneal injections of kisspeptin10 and kisspeptin234 (acting as a blocker) and were designated as RSA-KP10 and NP-KP234 groups. On the other hand, the control groups received intraperitoneal injections of normal saline (NS) and were referred to as RSA-NS and NP-NS groups. Each group comprised 6 mice, and uterine tissues from embryos at 9.5 days of gestation were meticulously collected for observation of embryo absorption and examination of the expression of the aforementioned proteins and genes. Results: The analysis revealed that the expression levels of kisspeptin, IGFBP1, Notch1, Akt, and Foxo1 were significantly lower in patients diagnosed with RSA compared to those in women with NP (P<0.01 for kisspeptin and P<0.05 for IGFBP1, Notch1, Akt, and Foxo1). After the introduction of kisspeptin10 to hESC, there was an observed enhancement in decidualization capability. Subsequently, the expression levels of Notch1, Akt, and Foxo1 showed an increase, but they decreased after interference with KISS1. Through immunofluorescence analysis, it was observed that proliferative hESC displayed a slender morphology, but they transitioned to a rounder and larger morphology post-decidualization. Concurrently, the expression of Notch1 increased, suggesting enhanced decidualization upon the administration of kisspeptin10, but the expression decreased after interference with KISS1. Further experimentation involved treating hESC with inhibitors specific to Notch1, Akt, and Foxo1 separately, revealing a regulatory sequence of Notch1/Akt/Foxo1 (P<0.05). In comparison to the NS group, NP mice administered with kisspeptin234 exhibited increased fetal absorption rates (P<0.001) and decreased expression of IGFBP1, Notch1, Akt, and Foxo1 (P<0.05). Conversely, RSA mice administered with kisspeptin10 demonstrated decreased fetal absorption rates (P<0.001) and increased expression levels of the aforementioned molecules (P<0.05). Conclusion: It is suggested that kisspeptin might exert its regulatory influence on the process of decidualization through the modulation of the Notch1/Akt/Foxo1 signaling cascade. A down-regulation of the expression levels of kisspeptin could result in suboptimal decidualization, which in turn might contribute to the development or progression of RSA.


Assuntos
Aborto Habitual , Decídua , Endométrio , Kisspeptinas , Proteínas Proto-Oncogênicas c-akt , Receptor Notch1 , Transdução de Sinais , Adulto , Feminino , Humanos , Gravidez , Aborto Habitual/metabolismo , Aborto Habitual/genética , Proliferação de Células , Decídua/metabolismo , Decídua/citologia , Endométrio/metabolismo , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Kisspeptinas/metabolismo , Kisspeptinas/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor Notch1/metabolismo , Receptor Notch1/genética
13.
Medicine (Baltimore) ; 103(28): e38866, 2024 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-38996103

RESUMO

BACKGROUND: This study aimed to explore the potential influence of kisspeptin (KISS1) levels on the etiology of placenta previa for early pregnancy diagnosis. METHODS: The study included 20 pregnant women diagnosed with placenta previa and 20 pregnant woman with normal pregnancies between 2021 and 2022. Plasma KISS1 levels were determined through biochemical analysis, while genetic analysis assessed KISS1 and KISS1 receptor gene expression levels. Immunohistochemical methods were employed to determine placenta KISS1 levels. RESULTS: The evaluation of KISS1 concentration in serum revealed a significant decrease in the placenta previa group compared to the control group (P < .001). KISS1 gene expression level 0.043-fold decreased in the placenta previa group (P < .001). Furthermore, the KISS1 receptor gene expression level increased 170-fold in the placenta previa group. CONCLUSIONS: Results from biochemical, immunohistochemical, and genetic analyses consistently indicated significantly reduced KISS1 expression in patients with placenta previa. These findings suggest a potential link between diminished KISS1 levels and the occurrence of placenta previa. KISS1 may play a critical role in the etiology of placenta previa. Detailed studies on angiogenesis, cell migration and tissue modeling should be conducted to understand possible mechanisms.


Assuntos
Kisspeptinas , Placenta Prévia , Humanos , Kisspeptinas/genética , Kisspeptinas/metabolismo , Feminino , Gravidez , Placenta Prévia/metabolismo , Adulto , Receptores de Kisspeptina-1/genética , Receptores de Kisspeptina-1/metabolismo , Placenta/metabolismo , Expressão Gênica
14.
PLoS Comput Biol ; 20(7): e1011820, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39083544

RESUMO

The pulsatile activity of gonadotropin-releasing hormone neurons (GnRH neurons) is a key factor in the regulation of reproductive hormones. This pulsatility is orchestrated by a network of neurons that release the neurotransmitters kisspeptin, neurokinin B, and dynorphin (KNDy neurons), and produce episodic bursts of activity driving the GnRH neurons. We show in this computational study that the features of coordinated KNDy neuron activity can be explained by a neural network in which connectivity among neurons is modular. That is, a network structure consisting of clusters of highly-connected neurons with sparse coupling among the clusters. This modular structure, with distinct parameters for intracluster and intercluster coupling, also yields predictions for the differential effects on synchronization of changes in the coupling strength within clusters versus between clusters.


Assuntos
Dinorfinas , Hormônio Liberador de Gonadotropina , Modelos Neurológicos , Rede Nervosa , Neurônios , Neurônios/fisiologia , Rede Nervosa/fisiologia , Animais , Dinorfinas/metabolismo , Dinorfinas/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Kisspeptinas/fisiologia , Neurocinina B/metabolismo , Neurocinina B/fisiologia , Biologia Computacional , Potenciais de Ação/fisiologia , Simulação por Computador , Humanos
15.
Reprod Fertil Dev ; 362024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38976640

RESUMO

Context There is mounting evidence implicating kisspeptin signalling in placental development and function. Aims This study aimed to elucidate kisspeptin's role in trophoblast invasion and migration using three experimental models. Methods First, we examined the mouse fetus and placenta in a kisspeptin receptor (Kiss1r) knockout (KO) model. Fetal/placental weights and gene expression (quantitative polymerase chain reaction) were assessed. Second, we determined kisspeptin effects on a human trophoblast (BeWo) cell line in vitro . Third, we examined KISS1 and KISS1R gene expression in human placenta from term and pre-term pregnancies. Key results No difference was found in fetal or placental weight between Kiss1r KO and wildtype mice. However, expression of the trophoblast invasion marker, Mmp2 mRNA, was greater in the placental labyrinth zone of Kiss1r KO mice. BeWo cell models of villus cytotrophoblast and syncytiotrophoblast cells exhibited kisspeptin protein expression, with greater expression in syncytiotrophoblast, consistent with KISS1 mRNA. Kisspeptin treatment inhibited the migratory potential of cytotrophoblast-like cells. Finally, while no difference was seen in KISS1 and KISS1R mRNA between term and pre-term placentas, we saw a difference in the relative expression of each gene pre-term. We also observed a positive correlation between KISS1 expression and maternal body mass index. Conclusions Our results indicate that kisspeptin may inhibit trophoblast invasion. Implications Further investigation is required to clarify specific regulatory mechanisms.


Assuntos
Movimento Celular , Kisspeptinas , Camundongos Knockout , Placenta , Receptores de Kisspeptina-1 , Trofoblastos , Kisspeptinas/metabolismo , Kisspeptinas/genética , Feminino , Trofoblastos/metabolismo , Receptores de Kisspeptina-1/metabolismo , Receptores de Kisspeptina-1/genética , Animais , Gravidez , Placenta/metabolismo , Movimento Celular/fisiologia , Humanos , Camundongos , Linhagem Celular , Placentação/fisiologia
16.
Methods Mol Biol ; 2811: 177-184, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39037658

RESUMO

KISS1 belongs to the family of metastasis suppressor genes. However, its role is not limited to blocking cancer metastasis. KISS1 and its by-product kisspeptins (KP) are important players in regulating the reproductive axis in different species and have new roles in controlling physiological balance and social behaviors. These diverse functions point to KISS1 as a potential therapeutic molecule. Here we describe a methodology to detect KISS1 and KP from cell lysate and conditioned media in cell lines. This will serve as a critical tool to study KISS1 processing in KP.


Assuntos
Kisspeptinas , Kisspeptinas/metabolismo , Humanos , Meios de Cultivo Condicionados , Linhagem Celular , Animais
17.
Domest Anim Endocrinol ; 89: 106868, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38901139

RESUMO

Lipopolysaccharide (LPS) from Gram-negative bacteria induces an immune response and impairs reproduction through suppression of gonadotropin releasing hormone (GnRH), subsequently luteinizing hormone (LH) secretion. While there is evidence that acute inflammation inhibits kisspeptin, little is known about the impact of chronic inflammation on this key reproductive neuropeptide in livestock species. Thus, we sought to examine a central mechanism whereby LPS suppresses LH secretion in sheep. Twenty wethers were randomly assigned to one of five treatment groups: control (CON; n=4), single acute IV LPS dose (SAD; n=4), daily acute IV LPS dose (DAD; n=4), daily increasing IV LPS dose (DID; n=4), and chronic subcutaneous LPS dose (CSD; n=4). On Days 1 and 7, blood samples were collected every 12 minutes for 360 minutes using jugular venipuncture. Following blood collection on Day 7, all animals were euthanized, brain tissue was perfused with 4% paraformaldehyde, and hypothalamic blocks were removed and processed for immunohistochemistry. On Day 1, LH pulse frequency was significantly lower (p=0.02) in SAD (0.25 ± 0.1 pulses/hour), DAD (0.25 ± 0.1 pulses/hour), DID (0.35 ± 0.1 pulses/hour), and CSD (0.40 ± 0.1 pulses/hour) compared to CON (0.70 ±0.1 pulses/hour). On Day 7, only DID animals (0.35 ± 0.1 pulses/hour) had significantly lower (p=0.049) LH pulse frequency compared to controls (0.85 ± 0.1 pulse/hour). Furthermore, only DID animals (33.3 ± 10.9 cells/section/animal) had significantly fewer (p=0.001) kisspeptin-immunopositive cells compared to controls (82.6 ± 13.6 cells/section/animal). Taken together, we suggest that daily increasing doses of LPS is a powerful inhibitor of kisspeptin neurons in young male sheep and a physiologically relevant model to examine the impact of chronic inflammation on the reproductive axis in livestock.


Assuntos
Inflamação , Kisspeptinas , Lipopolissacarídeos , Hormônio Luteinizante , Animais , Kisspeptinas/genética , Kisspeptinas/metabolismo , Masculino , Ovinos , Lipopolissacarídeos/farmacologia , Hormônio Luteinizante/sangue , Inflamação/veterinária , Núcleo Arqueado do Hipotálamo/metabolismo , Doenças dos Ovinos/metabolismo , Doenças dos Ovinos/induzido quimicamente , Regulação da Expressão Gênica/efeitos dos fármacos , Doença Crônica
18.
Placenta ; 154: 49-59, 2024 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-38878622

RESUMO

INTRODUCTION: Gestational diabetes mellitus (GDM) is a major pregnancy metabolic disorder and is strongly linked with obesity. Kisspeptin is a hormone that increases several thousand-fold in the maternal circulation during human pregnancy, with placenta as its main source. Studies have suggested that kisspeptin regulates trophoblast invasion and promotes pancreatic insulin secretion and peripheral insulin sensitivity. METHODS: In a well-characterized cohort of pregnant South African women and molecular and histological techniques, this study explored the impact and interaction of maternal obesity and GDM on kisspeptin (KISS1) signalling in relation to placental morphology and maternal and neonatal parameters. RESULTS: We found that GDM had no effect on placental KISS1 and KISS1R (KISS1 receptor) mRNA and/or protein expression. However, obesity reduced placental KISS1R mRNA expression even though overall KISS1 protein abundance or localization was not different from the non-obese group. Maternal and cord circulating KISS1 concentrations did not vary with obesity or GDM, but maternal circulating KISS1 was positively correlated with placenta weight in non-GDM obese women, and negatively correlated with placental intervillous space volume in non-GDM non-obese women. Cord serum KISS1 was positively correlated with infant weight in GDM obese women, but negatively correlated with maternal BMI in the non-obese GDM group. Placental syncytiotrophoblast extracellular vesicles exhibited detectable KISS1 and its abundance was ∼50 % lower in those from obese GDM compared to non-GDM women. DISCUSSION: This study shows maternal obesity and GDM can modulate placental kisspeptin signalling and placental morphological development with potential pathophysiological implications for clinically-relevant pregnancy and perinatal outcomes.


Assuntos
Diabetes Gestacional , Kisspeptinas , Obesidade , Placenta , Receptores de Kisspeptina-1 , Transdução de Sinais , Humanos , Feminino , Gravidez , Kisspeptinas/metabolismo , Placenta/metabolismo , Placenta/patologia , Diabetes Gestacional/metabolismo , Diabetes Gestacional/patologia , Adulto , África do Sul/epidemiologia , Obesidade/metabolismo , Obesidade/patologia , Receptores de Kisspeptina-1/metabolismo , Receptores de Kisspeptina-1/genética , Obesidade Materna/metabolismo
19.
FASEB J ; 38(13): e23743, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38877852

RESUMO

Kisspeptin signaling regulates energy homeostasis. Adiposity is the principal source and receiver of peripheral Kisspeptin, and adipose Kiss1 metastasis suppressor (Kiss1) gene expression is stimulated by exercise. However, whether the adipose Kiss1 gene regulates energy homeostasis and plays a role in adaptive alterations during prolonged exercise remains unknown. Here, we investigated the role of Kiss1 role in mice and adipose tissues and the adaptive changes it induces after exercise, using adipose-specific Kiss1 knockout (Kiss1adipoq-/-) and adeno-associated virus-induced adipose tissue Kiss1-overexpressing (Kiss1adipoq over) mice. We found that adipose-derived kisspeptin signal regulates lipid and glucose homeostasis to maintain systemic energy homeostasis, but in a sex-dependent manner, with more pronounced metabolic changes in female mice. Kiss1 regulated adaptive alterations of genes and proteins in tricarboxylic acid (TCA) cycle and oxidative phosphorylation (OxPhos) pathways in female gWAT following prolonged aerobic exercise. We could further show that adipose Kiss1 deficiency leads to reduced peroxisome proliferator-activated receptor gamma co-activator 1 alpha (PGC-1α) protein content of soleus muscle and maximum oxygen uptake (VO2 max) of female mice after prolonged exercise. Therefore, adipose Kisspeptin may be a novel adipokine that increases organ sensitivity to glucose, lipids, and oxygen following exercise.


Assuntos
Tecido Adiposo , Metabolismo Energético , Homeostase , Kisspeptinas , Camundongos Knockout , Condicionamento Físico Animal , Animais , Kisspeptinas/metabolismo , Kisspeptinas/genética , Feminino , Camundongos , Condicionamento Físico Animal/fisiologia , Masculino , Tecido Adiposo/metabolismo , Camundongos Endogâmicos C57BL , Adaptação Fisiológica
20.
Biomed Khim ; 70(3): 176-179, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38940207

RESUMO

The effect of a synthetic analog of kisspeptin 1, a peptide involved in the regulation of the hypothalamicpituitary- gonadal (HPG) stress axis, on the cortisol level of Danio rerio fish was investigated. Kisspeptin 1 was administered at doses of 2 µg/kg and 8 µg/kg followed by resting for 1 h and 4 h. We found that kisspeptin at doses of 2 µg/kg and 8 µg/kg increased cortisol levels, with a significant spike in cortisol levels at 1 h post-injection.


Assuntos
Hidrocortisona , Kisspeptinas , Proteínas de Peixe-Zebra , Peixe-Zebra , Animais , Kisspeptinas/farmacologia , Kisspeptinas/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Masculino , Feminino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...