Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.092
Filtrer
1.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(3): 542-551, 2024 May 20.
Article de Chinois | MEDLINE | ID: mdl-38948287

RÉSUMÉ

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.


Sujet(s)
Avortements à répétition , Caduques , Endomètre , Protéine O1 à motif en tête de fourche , Kisspeptines , Protéines proto-oncogènes c-akt , Récepteur Notch1 , Transduction du signal , Femelle , Protéine O1 à motif en tête de fourche/métabolisme , Protéine O1 à motif en tête de fourche/génétique , Humains , Protéines proto-oncogènes c-akt/métabolisme , Endomètre/métabolisme , Caduques/métabolisme , Caduques/cytologie , Grossesse , Récepteur Notch1/métabolisme , Récepteur Notch1/génétique , Avortements à répétition/métabolisme , Avortements à répétition/génétique , Kisspeptines/métabolisme , Kisspeptines/génétique , Adulte , Protéine-1 de liaison aux IGF/métabolisme , Protéine-1 de liaison aux IGF/génétique , Prolifération cellulaire
2.
Reprod Domest Anim ; 59(6): e14630, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38847348

RÉSUMÉ

This study evaluated whether the treatment of pseudopregnancy in bitches with vitamin B6 modulates uterine expression of receptors for progesterone (PR), oestrogen (ERα), androgen (AR), thyroid hormone (TRα) and the kisspeptin/Kiss1r system. Eighteen pseudopregnant bitches were treated for 20 days in groups receiving placebo (n = 6); cabergoline (5 µg/kg/day; n = 6); or vitamin B6 (50 mg/kg/day; n = 6). Blood was collected on the 1st day of drug administration and 120 h later to measure serum prolactin (PRL). After treatment, they were ovariohysterectomized and uterine fragments were collected for histomorphometry and immunohistochemical evaluation of PR, ERα, AR, TRα, Kiss1 and Kiss1r. After 120 h of cabergoline or vitamin B6 treatment, PRL levels were reduced in the bitches, confirming the antiprolactinemic effect of these drugs. Furthermore, regardless of treatment, the animals exhibited uterine histomorphometry consistent with dioestrus. The PR showed strong immunostaining in all regions and an increase in scores was observed for this receptor in animals treated with vitamin B6 in deep glands. In contrast, ERα and Kiss1R receptors showed weak to no immunostaining in all uterine regions and no changes between groups. Regarding AR, most animals treated with vitamin B6 showed increased trends in the deep gland and myometrium marking scores. In contrast, in both vitamin B6 and cabergoline treatments, a reduction in TRα marking scores was observed compared to the control group. In addition, on the endometrial surface, a reduction was observed in the marked area of Kiss1 after administration of cabergoline when compared to the pseudopregnant control group. These findings shed valuable insight into the use of vitamin B6 as a drug with actions similar to cabergoline in reducing PRL and uterine modulation in bitches.


Sujet(s)
Cabergoline , Kisspeptines , Prolactine , Grossesse nerveuse , Utérus , Animaux , Femelle , Chiens , Kisspeptines/pharmacologie , Kisspeptines/métabolisme , Utérus/effets des médicaments et des substances chimiques , Utérus/métabolisme , Cabergoline/pharmacologie , Prolactine/métabolisme , Grossesse nerveuse/médecine vétérinaire , Grossesse nerveuse/métabolisme , Récepteurs à la progestérone/métabolisme , Récepteurs aux androgènes/métabolisme , Ergolines/pharmacologie
3.
Biomed Khim ; 70(3): 176-179, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38940207

RÉSUMÉ

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.


Sujet(s)
Hydrocortisone , Kisspeptines , Protéines de poisson-zèbre , Danio zébré , Animaux , Kisspeptines/pharmacologie , Kisspeptines/métabolisme , Protéines de poisson-zèbre/métabolisme , Mâle , Femelle
4.
FASEB J ; 38(13): e23743, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38877852

RÉSUMÉ

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.


Sujet(s)
Tissu adipeux , Métabolisme énergétique , Homéostasie , Kisspeptines , Souris knockout , Conditionnement physique d'animal , Animaux , Kisspeptines/métabolisme , Kisspeptines/génétique , Femelle , Souris , Conditionnement physique d'animal/physiologie , Mâle , Tissu adipeux/métabolisme , Souris de lignée C57BL , Adaptation physiologique
5.
Int J Mol Sci ; 25(12)2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38928237

RÉSUMÉ

The physiology of reproduction has been of interest to researchers for centuries. The purpose of this work is to review the development of our knowledge on the neuroendocrine background of the regulation of ovulation. We first describe the development of the pituitary gland, the structure of the median eminence (ME), the connection between the hypothalamus and the pituitary gland, the ovarian and pituitary hormones involved in ovulation, and the pituitary cell composition. We recall the pioneer physiological and morphological investigations that drove development forward. The description of the supraoptic-paraventricular magnocellular and tuberoinfundibular parvocellular systems and recognizing the role of the hypophysiotropic area were major milestones in understanding the anatomical and physiological basis of reproduction. The discovery of releasing and inhibiting hormones, the significance of pulse and surge generators, the pulsatile secretion of the gonadotropin-releasing hormone (GnRH), and the subsequent pulsatility of luteinizing (LH) and follicle-stimulating hormones (FSH) in the human reproductive physiology were truly transformative. The roles of three critical neuropeptides, kisspeptin (KP), neurokinin B (NKB), and dynorphin (Dy), were also identified. This review also touches on the endocrine background of human infertility and assisted fertilization.


Sujet(s)
Système neuroendocrinien , Ovulation , Humains , Ovulation/physiologie , Femelle , Système neuroendocrinien/physiologie , Système neuroendocrinien/métabolisme , Animaux , Hypophyse/métabolisme , Kisspeptines/métabolisme , Neurokinine B/métabolisme , Hormone lutéinisante/métabolisme , Hormone de libération des gonadotrophines/métabolisme , Dynorphines/métabolisme , Hypothalamus/métabolisme , Hypothalamus/physiologie
6.
Int J Mol Sci ; 25(9)2024 May 06.
Article de Anglais | MEDLINE | ID: mdl-38732265

RÉSUMÉ

Epigenetic alterations my play a role in the aggressive behavior of Non-Small Cell Lung Cancer (NSCLC). Treatment with the histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA, vorinostat) has been reported to interfere with the proliferative and invasive potential of NSCLC cells. In addition, the DNA methyltransferase inhibitor azacytidine (AZA, vidaza) can modulate the levels of the metastasis suppressor KiSS-1. Thus, since cisplatin is still clinically available for NSCLC therapy, the aim of this study was to evaluate drug combinations between cisplatin and SAHA as well as AZA using cisplatin-sensitive H460 and -resistant H460/Pt NSCLC cells in relation to KiSS-1 modulation. An analysis of drug interaction according to the Combination-Index values indicated a more marked synergistic effect when the exposure to SAHA or AZA preceded cisplatin treatment with respect to a simultaneous schedule. A modulation of proteins involved in apoptosis (p53, Bax) was found in both sensitive and resistant cells, and compared to the treatment with epigenetic agents alone, the combination of cisplatin and SAHA or AZA increased apoptosis induction. The epigenetic treatments, both as single agents and in combination, increased the release of KiSS-1. Finally, the exposure of cisplatin-sensitive and -resistant cells to the kisspeptin KP10 enhanced cisplatin induced cell death. The efficacy of the combination of SAHA and cisplatin was tested in vivo after subcutaneous inoculum of parental and resistant cells in immunodeficient mice. A significant tumor volume inhibition was found when mice bearing advanced tumors were treated with the combination of SAHA and cisplatin according to the best schedule identified in cellular studies. These results, together with the available literature, support that epigenetic drugs are amenable for the combination treatment of NSCLC, including patients bearing cisplatin-resistant tumors.


Sujet(s)
Azacitidine , Cisplatine , Résistance aux médicaments antinéoplasiques , Épigenèse génétique , Kisspeptines , Tumeurs du poumon , Vorinostat , Cisplatine/pharmacologie , Animaux , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Humains , Souris , Épigenèse génétique/effets des médicaments et des substances chimiques , Kisspeptines/métabolisme , Kisspeptines/pharmacologie , Kisspeptines/génétique , Lignée cellulaire tumorale , Vorinostat/pharmacologie , Azacitidine/pharmacologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Tests d'activité antitumorale sur modèle de xénogreffe , Antinéoplasiques/pharmacologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Synergie des médicaments , Inhibiteurs de désacétylase d'histone/pharmacologie , Femelle
7.
Mol Reprod Dev ; 91(5): e23744, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38800960

RÉSUMÉ

This study unravels the intricate interplay between photoperiod, melatonin, and kisspeptin to orchestrate the pubertal onset of Common carp. Female fingerlings exposed to long days (LD) exhibited a hormonal crescendo, with upregulated hypothalamic-pituitary-ovarian (HPO) axis genes (kiss1, kiss1r, kiss2, gnrh2, gnrh3) and their downstream targets (lhr, fshr, ar1, esr1). However, the expression of the melatonin receptor (mtnr1a) diminished in LD, suggesting a potential inhibitory role. This hormonal symphony was further amplified by increased activity of key transcriptional regulators (gata1, gata2, cdx1, sp1, n-myc, hoxc8, plc, tac3, tacr3) and decreased expression of delayed puberty genes (mkrn1, dlk1). In contrast, short days (SD) muted this hormonal chorus, with decreased gnrh gene and regulator expression, elevated mtnr1a, and suppressed gonadal development. In in-vitro, estradiol mimicked the LD effect, boosting gnrh and regulator genes while dampening mtnr1a and melatonin-responsive genes. Conversely, melatonin acted as a conductor, downregulating gnrh and regulator genes and amplifying mtnr1a. Our findings illuminate the crucial roles of melatonin and kisspeptin as opposing forces in regulating pubertal timing. LD-induced melatonin suppression allows the kisspeptin symphony to flourish, triggering GnRH release and, ultimately, gonadal maturation. This delicate dance between photoperiod, melatonin, and kisspeptin orchestrates common carp's transition from juvenile to reproductive life.


Sujet(s)
Carpes (poisson) , Kisspeptines , Mélatonine , Photopériode , Maturation sexuelle , Animaux , Mélatonine/métabolisme , Kisspeptines/métabolisme , Kisspeptines/génétique , Femelle , Carpes (poisson)/métabolisme , Carpes (poisson)/génétique , Carpes (poisson)/croissance et développement , Carpes (poisson)/physiologie , Maturation sexuelle/physiologie , Protéines de poisson/métabolisme , Protéines de poisson/génétique
8.
Sci Rep ; 14(1): 10190, 2024 05 03.
Article de Anglais | MEDLINE | ID: mdl-38702366

RÉSUMÉ

Dysfunction of central serotonergic neurons is known to cause depressive disorders in humans, who often show reproductive and/or glucose metabolism disorders. This study examined whether dorsal raphe (DR) serotonergic neurons sense high glucose availability to upregulate reproductive function via activating hypothalamic arcuate (ARC) kisspeptin neurons (= KNDy neurons), a dominant stimulator of gonadotropin-releasing hormone (GnRH)/gonadotropin pulses, using female rats and goats. RNA-seq and histological analysis revealed that stimulatory serotonin-2C receptor (5HT2CR) was mainly expressed in the KNDy neurons in female rats. The serotonergic reuptake inhibitor administration into the mediobasal hypothalamus (MBH), including the ARC, significantly blocked glucoprivic suppression of luteinizing hormone (LH) pulses and hyperglycemia induced by intravenous 2-deoxy-D-glucose (2DG) administration in female rats. A local infusion of glucose into the DR significantly increased in vivo serotonin release in the MBH and partly restored LH pulses and hyperglycemia in the 2DG-treated female rats. Furthermore, central administration of serotonin or a 5HT2CR agonist immediately evoked GnRH pulse generator activity, and central 5HT2CR antagonism blocked the serotonin-induced facilitation of GnRH pulse generator activity in ovariectomized goats. These results suggest that DR serotonergic neurons sense high glucose availability to reduce gluconeogenesis and upregulate reproductive function by activating GnRH/LH pulse generator activity in mammals.


Sujet(s)
Glucose , Capra , Hormone de libération des gonadotrophines , Hormone lutéinisante , Récepteur de la sérotonine de type 5-HT2C , Neurones sérotonergiques , Animaux , Hormone lutéinisante/métabolisme , Femelle , Récepteur de la sérotonine de type 5-HT2C/métabolisme , Rats , Neurones sérotonergiques/métabolisme , Hormone de libération des gonadotrophines/métabolisme , Glucose/métabolisme , Sérotonine/métabolisme , Kisspeptines/métabolisme , Noyau arqué de l'hypothalamus/métabolisme , Noyau arqué de l'hypothalamus/effets des médicaments et des substances chimiques , Noyau dorsal du raphé/métabolisme , Noyau dorsal du raphé/effets des médicaments et des substances chimiques , Rat Sprague-Dawley
9.
Metabolism ; 157: 155932, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38729600

RÉSUMÉ

BACKGROUND: Obesity-induced hypogonadism (OIH) is a prevalent, but often neglected condition in men, which aggravates the metabolic complications of overweight. While hypothalamic suppression of Kiss1-encoded kisspeptin has been suggested to contribute to OIH, the molecular mechanisms for such repression in obesity, and the therapeutic implications thereof, remain unknown. METHODS: A combination of bioinformatic, expression and functional analyses was implemented, assessing the role of the evolutionary-conserved miRNAs, miR-137 and miR-325, in mediating obesity-induced suppression of hypothalamic kisspeptin, as putative mechanism of central hypogonadism and metabolic comorbidities. The implications of such miR-137/325-kisspeptin interplay for therapeutic intervention in obesity were also explored using preclinical OIH models. RESULTS: MiR-137/325 repressed human KISS1 3'-UTR in-vitro and inhibited hypothalamic kisspeptin content in male rats, while miR-137/325 expression was up-regulated, and Kiss1/kisspeptin decreased, in the medio-basal hypothalamus of obese rats. Selective over-expression of miR-137 in Kiss1 neurons reduced Kiss1/ kisspeptin and partially replicated reproductive and metabolic alterations of OIH in lean mice. Conversely, interference of the repressive actions of miR-137/325 selectively on Kiss1 3'-UTR in vivo, using target-site blockers (TSB), enhanced kisspeptin content and reversed central hypogonadism in obese rats, together with improvement of glucose intolerance, insulin resistance and cardiovascular and inflammatory markers, despite persistent exposure to obesogenic diet. Reversal of OIH by TSB miR-137/325 was more effective than chronic kisspeptin or testosterone treatments in obese rats. CONCLUSIONS: Our data disclose that the miR-137/325-Kisspeptin repressive interaction is a major player in the pathogenesis of obesity-induced hypogonadism and a putative druggable target for improved management of this condition and its metabolic comorbidities in men suffering obesity. SIGNIFICANCE STATEMENT: Up to half of the men suffering obesity display also central hypogonadism, an often neglected complication of overweight that can aggravate the clinical course of obesity and its complications. The mechanisms for such obesity-induced hypogonadism remain poorly defined. We show here that the evolutionary conserved miR137/miR325 tandem centrally mediates obesity-induced hypogonadism via repression of the reproductive-stimulatory signal, kisspeptin; this may represent an amenable druggable target for improved management of hypogonadism and other metabolic complications of obesity.


Sujet(s)
Hypogonadisme , Hypothalamus , Kisspeptines , microARN , Obésité , microARN/génétique , microARN/métabolisme , Hypogonadisme/génétique , Hypogonadisme/métabolisme , Hypogonadisme/complications , Kisspeptines/génétique , Kisspeptines/métabolisme , Animaux , Obésité/métabolisme , Obésité/complications , Obésité/génétique , Mâle , Rats , Hypothalamus/métabolisme , Humains , Souris , Rat Wistar , Comorbidité
10.
Behav Brain Res ; 468: 115035, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38703793

RÉSUMÉ

Parkinson's Disease is a progressive neurodegenerative disorder characterized by motor symptoms resulting from the loss of nigrostriatal dopaminergic neurons. Kisspeptins (KPs) are a family of neuropeptides that are encoded by the Kiss-1 gene, which exert their physiological effects through interaction with the GPR54 receptor. In the current investigation, we investigated the prospective protective effects of central KP-54 treatments on nigrostriatal dopaminergic neurons and consequent motor performance correlates in 6-hydroxydopamine (6-OHDA)-lesioned rats. Male adult Sprague Dawley rats underwent stereotaxic injection of 6-OHDA into the right medial forebrain bundle to induce hemiparkinsonism. Following surgery, rats received chronic central treatments of nasal or intracerebroventricular KP-54 (logarithmically increasing doses) for seven consecutive days. Motor performance was evaluated seven days post-surgery utilizing the open field test and catalepsy test. The levels of dopamine in the striatum were determined with mass spectrometry. Immunohistochemical analysis was conducted to assess the immunoreactivities of tyrosine hydroxylase (TH) and the GPR54 in the substantia nigra. The dose-response curve revealed a median effective dose value of ≈3 nmol/kg for both central injections. Due to its non-invasive and effective nature, nasal administration was utilized in the second phase of our study. Chronic administration of KP-54 (3nmol/kg, nasally) significantly protected 6-OHDA-induced motor deficits. Nasal KP-54 attenuated the loss of nigrostriatal dopaminergic neurons induced by 6-OHDA. Additionally, significant correlations were observed between motor performance and nigrostriatal dopamine levels. Immunohistochemical analysis demonstrated the localization of the GPR54 within TH-positive nigral cells. These findings suggest the potential efficacy of central KP-54 on motor impairments in hemiparkinsonism.


Sujet(s)
Administration par voie nasale , Corps strié , Dopamine , Neurones dopaminergiques , Kisspeptines , Oxidopamine , Syndromes parkinsoniens , Rat Sprague-Dawley , Substantia nigra , Animaux , Mâle , Substantia nigra/effets des médicaments et des substances chimiques , Substantia nigra/métabolisme , Dopamine/métabolisme , Oxidopamine/pharmacologie , Rats , Corps strié/effets des médicaments et des substances chimiques , Corps strié/métabolisme , Kisspeptines/administration et posologie , Kisspeptines/pharmacologie , Kisspeptines/métabolisme , Syndromes parkinsoniens/traitement médicamenteux , Syndromes parkinsoniens/métabolisme , Neurones dopaminergiques/effets des médicaments et des substances chimiques , Neurones dopaminergiques/métabolisme , Modèles animaux de maladie humaine , Activité motrice/effets des médicaments et des substances chimiques , Tyrosine 3-monooxygenase/métabolisme
11.
J Recept Signal Transduct Res ; 44(1): 19-26, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38647103

RÉSUMÉ

Kisspeptin is an important hormone involved in the stimulation of the hypothalamo-pituitary gonadal (HPG) axis. The HPG axis can be suppressed in certain conditions such as stress, which gives rise to the activation of the hypothalamo-pituitary-adrenal (HPA) axis. However, the physiological role of kisspeptin in the interaction of HPG and HPA axis is not fully understood yet. This study was conducted to investigate the possible effects of central kisspeptin injection on HPG axis as well as HPA axis activity. Adult male Wistar rats were randomly divided into seven groups as followed: sham (control), kisspeptin (50 pmol), P234 (1 nmol), kisspeptin + p234, kisspeptin + antalarmin (0.1 µg), kisspeptin + astressin 2B (1 µg), and kisspeptin + atosiban (300 ng/rat) (n = 10 each group). At the end of the experiments, the hypothalamus, pituitary, and serum samples of the rats were collected. There was no significant difference in corticotropic-releasing hormone immunoreactivity in the paraventricular nucleus of the hypothalamus, serum adrenocorticotropic hormone, and corticosterone levels among all groups. Moreover, no significant difference was detected in pituitary oxytocin level. Serum follicle-stimulating hormone and luteinizing hormone levels of the kisspeptin, kisspeptin + antalarmin, and kisspeptin + astressin 2B groups were significantly higher than the control group. Serum testosterone levels were significantly higher in the kisspeptin kisspeptin + antalarmin, kisspeptin + astressin 2B, and kisspeptin + atosiban groups compared to the control group. Our findings suggest that central kisspeptin injection causes activation in the HPG axis, but not the HPA axis in male rats.


Sujet(s)
Axe hypothalamohypophysaire , Kisspeptines , Axe hypophyso-surrénalien , Rat Wistar , Animaux , Mâle , Kisspeptines/administration et posologie , Kisspeptines/pharmacologie , Kisspeptines/métabolisme , Axe hypothalamohypophysaire/effets des médicaments et des substances chimiques , Axe hypothalamohypophysaire/métabolisme , Axe hypophyso-surrénalien/effets des médicaments et des substances chimiques , Axe hypophyso-surrénalien/métabolisme , Rats , Fragments peptidiques/administration et posologie , Hormone lutéinisante/sang , Hormone lutéinisante/métabolisme , Corticostérone/sang , Vasotocine/pharmacologie , Vasotocine/administration et posologie , Testostérone/sang , Injections ventriculaires , Gonades/métabolisme , Gonades/effets des médicaments et des substances chimiques , Hypophyse/métabolisme , Hypophyse/effets des médicaments et des substances chimiques , Hormone de libération des gonadotrophines/métabolisme , Hormone corticotrope/sang , Corticolibérine , Oligopeptides
12.
Domest Anim Endocrinol ; 88: 106850, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38640803

RÉSUMÉ

Kisspeptins are neuropeptides encoded by the Kiss1 gene that was discovered as a metastasis suppressor gene in melanoma and breast cancer. Kisspeptin has pivotal functions for gonadotropin-releasing hormone secretion and plays integrated roles in the hypothalamic-pituitary-gonadal axis. However, little is known about the peripheral expression of kisspeptin in ruminants, especially in the female reproductive tract. Here, the objectives of the current study were to investigate the spatial localization of kisspeptin and mRNA expression of Kiss1 and its receptor (Kiss1r) in the fallopian tubes (FT) and uterus of goats at varied reproductive activity (cyclic versus true anoestrous goats, n=6, each). Specimens of the uterus and FT were collected and fixed using paraformaldehyde to investigate the localizations of kisspeptin in the selected tissues by immunohistochemistry. Another set of samples was snape-frozen to identify the expressions of mRNAs encoding Kiss1 and Kiss1r using real-time PCR. Results revealed immunolocalizations of kisspeptin in the uterus and the FT. The staining of kisspeptin was found mainly in the mucosal epithelium of the uterus the FT, and the endometrial glands. Very intense staining of kisspeptin was found in the uterine and FT specimens in the true anoestrous goats compared to that in cyclic ones. The expression of mRNA encoding Kiss1 gene was significantly higher in the uterine specimen of cyclic goats (1.00±0.09) compared to that in the true anoestrous goats (0.62±0.08) (P ˂0.05), while the expression of mRNA encoding Kiss1r was significantly (P ˂0.001) higher in the uterine tissues of true anoestrous goats (1.78±0.17) compared to that in cyclic ones (1.00±0.11). In conclusion, immunohistochemical localization of kisspeptin and the expression of mRNA encoding Kiss1/Kiss1r revealed spatial changes in the uterus and FT of goats according to the reproductive potential of goats (cyclic versus true anoestrous goats). However, the definitive local role of kisspeptin in the uterus and FT need further investigation.


Sujet(s)
Trompes utérines , Capra , Kisspeptines , Utérus , Animaux , Femelle , Capra/physiologie , Capra/génétique , Capra/métabolisme , Kisspeptines/génétique , Kisspeptines/métabolisme , Utérus/métabolisme , Trompes utérines/métabolisme , Trompes utérines/composition chimique , ARN messager/métabolisme , ARN messager/génétique , Reproduction/physiologie , Régulation de l'expression des gènes/physiologie , Récepteur de la Kisspeptine-1/génétique , Récepteur de la Kisspeptine-1/métabolisme , Anoestrus/métabolisme
13.
Sci Rep ; 14(1): 8229, 2024 04 08.
Article de Anglais | MEDLINE | ID: mdl-38589425

RÉSUMÉ

Polycystic ovary syndrome (PCOS) is the most common endocrine disorder affecting 5-20% of reproductive-age women. However, the treatment of PCOS is mainly based on symptoms and not on its pathophysiology. Neuroendocrine disturbance, as shown by an elevated LH/FSH ratio in PCOS patients, was thought to be the central mechanism of the syndrome, especially in lean PCOS. LH and FSH secretion are influenced by GnRH pulsatility of GnRH neurons in the hypothalamus. Kisspeptin is the main regulator of GnRH secretion, whereas neurokinin B (NKB) and dynorphin regulate kisspeptin secretion in KNDy neurons. This study aims to deepen the understanding of the neuroendocrine disorder in lean PCOS patients and its potential pathophysiology-based therapy. A cross-sectional study was performed at Dr. Cipto Mangunkusumo Kencana Hospital and the IMERI UI HRIFP cluster with 110 lean PCOS patients as subjects. LH, FSH, LH/FSH ratio, kisspeptin, NKB, dynorphin, leptin, adiponectin, AMH, fasting blood glucose, fasting insulin, HOMA-IR, testosterone, and SHBG were measured. Bivariate and path analyses were performed to determine the relationship between variables. There was a negative association between dynorphin and kisspeptin, while NKB levels were not associated with kisspeptin. There was no direct association between kisspeptin and the LH/FSH ratio; interestingly, dynorphin was positively associated with the LH/FSH ratio in both bivariate and pathway analyses. AMH was positively correlated with the LH/FSH ratio in both analyses. Path analysis showed an association between dynorphin and kisspeptin levels in lean PCOS, while NKB was not correlated with kisspeptin. Furthermore, there was a correlation between AMH and the LH/FSH ratio, but kisspeptin levels did not show a direct significant relationship with the LH/FSH ratio. HOMA-IR was negatively associated with adiponectin levels and positively associated with leptin and FAI levels. In conclusion, AMH positively correlates with FAI levels and is directly associated with the LH/FSH ratio, showing its important role in neuroendocrinology in lean PCOS. From the path analysis, AMH was also an intermediary variable between HOMA-IR and FAI with the LH/FSH ratio. Interestingly, this study found a direct positive correlation between dynorphin and the LH/FSH ratio, while no association between kisspeptin and the LH/FSH ratio was found. Further research is needed to investigate AMH and dynorphin as potential therapeutic targets in the management of lean PCOS patients.


Sujet(s)
Hormone lutéinisante , Syndrome des ovaires polykystiques , Femelle , Humains , Dynorphines/métabolisme , Leptine , Kisspeptines/métabolisme , Études transversales , Adiponectine , Neurokinine B/métabolisme , Hormone de libération des gonadotrophines/métabolisme , Hormone folliculostimulante
14.
Gen Comp Endocrinol ; 353: 114528, 2024 07 01.
Article de Anglais | MEDLINE | ID: mdl-38643848

RÉSUMÉ

Kisspeptin is a peptide that plays an important role through its effects on the hypothalamus-pituitary-gonadal (HPG) axis. It has also been implicated in sexual behavior. The present study investigated whether the relationship between kisspeptin and sexual behavior is independent of the HPG axis, i.e., testosterone. Sexual behavior was examined after the administration of kisspeptin to gonadally intact male rats and gonadectomized male rats that received testosterone supplementation. Other male rats were also observed for sexual behavior once a week from 2 to 5 weeks after gonadectomy and receiving kisspeptin for the sixth postoperative week. Sexual behavior in female rats serving as the partner for each male was also observed. Female rats were not administered kisspeptin in the present study. The results obtained showed that the administration of kisspeptin increased precopulatory behavior in gonadally intact male rats and gonadectomized male rats that received testosterone supplementation and proceptive behavior in their female partners. Precopulatory behavior in males and receptive behavior in females increased, while copulatory behavior in males and receptive behavior in females remained unchanged. Furthermore, the administration of kisspeptin increased precopulatory behavior in gonadectomized males, but did not affect receptive behavior in females. These results suggest that kisspeptin affected males independently and/or supplementally to testosterone, and also that changes in the presence of testosterone in males had an impact on proceptive behavior in their female partners. In conclusion, kisspeptin may involve an as-yet-unidentified neural pathway in sexual desire independently of the HPG axis.


Sujet(s)
Kisspeptines , Comportement sexuel chez les animaux , Testostérone , Animaux , Kisspeptines/métabolisme , Kisspeptines/pharmacologie , Mâle , Testostérone/pharmacologie , Femelle , Rats , Comportement sexuel chez les animaux/effets des médicaments et des substances chimiques , Comportement sexuel chez les animaux/physiologie , Rat Wistar , Copulation/effets des médicaments et des substances chimiques , Copulation/physiologie
15.
Cell Mol Biol (Noisy-le-grand) ; 70(3): 254-259, 2024 Mar 31.
Article de Anglais | MEDLINE | ID: mdl-38650158

RÉSUMÉ

We attempted to clarify clinical value of KiSS-1 and MMP-2 levels in breast cancer (BC) tissue in evaluating prognosis of elderly BC patients after modified radical mastectomy (MCM). The data of 192 elderly female BC patients receiving MCM in our hospital from January 2018 to December 2022 were collected. According to prognosis, patients received division into poor prognosis group (n = 43) and good prognosis group (n = 149). The serum CEA level and KiSS-1 and MMP-2 levels in BC tissue received measurement in both groups. The predictive value of KiSS-1 and MMP-2 alone and jointly in adverse prognosis of elderly BC patients after MCM received assessment. Results showed that No statistical significance was exhibited between both groups in general data (P > 0.05). The serum CEA level and MMP-2 expression in BC tissue in poor prognosis group exhibited elevation relative to those in good prognosis group, and KiSS-1 expression in BC tissue in poor prognosis group exhibited depletion relative to that in good prognosis group, indicating statistical significance (P < 0.05). The high-level KiSS-1 might be a protective element for adverse prognosis of elderly BC patients after MCM, and high-level CEA and MMP-2 might be an independent risk element for adverse prognosis of elderly BC patients after MCM (P < 0.05). KiSS-1 and MMP-2 alone and jointly predicted AUC of adverse prognosis in elderly BC patients after MCM were 0.93, 0.802 and 0.958, with certain predictive values; when cutoff values of KiSS-1 and MMP-2 were 6.15 and 2.26, the predictive value was the best. In conclusion, KiSS-1 and MMP-2 levels in BC tissue possess relation to adverse prognosis of MCM. KiSS-1 and MMP-2 levels in elderly BC patients before surgery may be detected in the future to assist in prognosis evaluation of elderly BC patients after MCM.


Sujet(s)
Tumeurs du sein , Kisspeptines , Mastectomie radicale modifiée , Matrix metalloproteinase 2 , Humains , Femelle , Matrix metalloproteinase 2/métabolisme , Matrix metalloproteinase 2/sang , Tumeurs du sein/chirurgie , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/mortalité , Sujet âgé , Pronostic , Kisspeptines/métabolisme , Courbe ROC , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/sang , Sujet âgé de 80 ans ou plus
16.
J Diabetes ; 16(4): e13541, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38599822

RÉSUMÉ

Kisspeptins (KPs) are proteins that were first recognized to have antimetastatic action. Later, the critical role of this peptide in the regulation of reproduction was proved. In recent years, evidence has been accumulated supporting a role for KPs in regulating metabolic processes in a sexual dimorphic manner. It has been proposed that KPs regulate metabolism both indirectly via gonadal hormones and/or directly via the kisspeptin receptor in the brain, brown adipose tissue, and pancreas. The aim of the review is to provide both experimental and clinical evidence indicating that KPs are peptides linking metabolism and reproduction. We propose that KPs could be used as a potential target to treat both metabolic and reproductive abnormalities. Thus, we focus on the consequences of disruptions in KPs and their receptors in metabolic conditions such as diabetes, undernutrition, obesity, and reproductive disorders (hypogonadotropic hypogonadism and polycystic ovary syndrome). Data from both animal models and human subjects indicate that alterations in KPs in the case of metabolic imbalance lead also to disruptions in reproductive functions. Changes both in the hypothalamic and peripheral KP systems in animal models of the aforementioned disorders are discussed. Finally, an overview of current clinical studies involving KP in fertility and metabolism show fewer studies on metabolism (15%) and only one to date on both. Presented data indicate a dynamic and emerging field of KP studies as possible therapeutic targets in treatments of both reproductive and metabolic dysfunctions.


Sujet(s)
Kisspeptines , Reproduction , Animaux , Femelle , Humains , Kisspeptines/métabolisme , Hypothalamus/métabolisme , Obésité/métabolisme , Peptides
17.
Am J Respir Cell Mol Biol ; 70(6): 507-518, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38512807

RÉSUMÉ

Airway remodeling is a cardinal feature of asthma, associated with increased airway smooth muscle (ASM) cell mass and upregulation of extracellular matrix deposition. Exaggerated ASM cell migration contributes to excessive ASM mass. Previously, we demonstrated the alleviating role of Kp (kisspeptin) receptor (KISS1R) activation by Kp-10 in mitogen (PDGF [platelet-derived growth factor])-induced human ASM cell proliferation in vitro and airway remodeling in vivo in a mouse model of asthma. Here, we examined the mechanisms by which KISS1R activation regulates mitogen-induced ASM cell migration. KISS1R activation using Kp-10 significantly inhibited PDGF-induced ASM cell migration, further confirmed using KISS1R shRNA. Furthermore, KISS1R activation modulated F/G actin dynamics and the expression of promigration proteins like CDC42 (cell division control protein 42) and cofilin. Mechanistically, we observed reduced ASM RhoA-GTPAse with KISS1R activation. The antimigratory effect of KISS1R was abolished by PKA (protein kinase A)-inhibitory peptide. Conversely, KISS1R activation significantly increased cAMP and phosphorylation of CREB (cAMP-response element binding protein) in PDGF-exposed ASM cells. Overall, these results highlight the alleviating properties of Kp-10 in the context of airway remodeling.


Sujet(s)
Mouvement cellulaire , Kisspeptines , Myocytes du muscle lisse , Facteur de croissance dérivé des plaquettes , Récepteur de la Kisspeptine-1 , Transduction du signal , Protéine G RhoA , Humains , Mouvement cellulaire/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Kisspeptines/métabolisme , Facteur de croissance dérivé des plaquettes/métabolisme , Facteur de croissance dérivé des plaquettes/pharmacologie , Récepteur de la Kisspeptine-1/métabolisme , Récepteur de la Kisspeptine-1/génétique , Protéine G RhoA/métabolisme , Récepteurs couplés aux protéines G/métabolisme , Protéine G cdc42/métabolisme , Remodelage des voies aériennes , AMP cyclique/métabolisme , Cyclic AMP-Dependent Protein Kinases/métabolisme , Cellules cultivées , Facteurs de dépolymérisation de l'actine/métabolisme , Actines/métabolisme , Prolifération cellulaire
18.
Theriogenology ; 219: 157-166, 2024 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-38432143

RÉSUMÉ

To understand better the role that kisspeptin plays in regulating seasonal and estrous cycle changes in the mare, this study investigated the number, location and interactions between GnRH, kisspeptin and RFRP-3 neurons in the equine hypothalamus. Hypothalami were collected from mares during the non-breeding season, vernal transition and various stages of the breeding season. Fluorescent immunohistochemistry was used to label the neuropeptides of interest. GnRH cells were observed primarily in the arcuate nucleus (ARC), while very few labeled cells were identified in the pre-optic area (POA). Kisspeptin cells were identified primarily in the ARC, with a small number of cells observed dorsal to the ARC, surrounding the third ventricle (3V). The mean number of kisspeptin cells varied between animals and typically showed no pattern associated with season or stage of estrous cycle, but a seasonal difference was identified in the ARC population. Small numbers of RFRP-3 cells were observed in the ARC, ventromedial hypothalamus (VMH) and dorsomedial hypothalamus (DMH). The mean number of RFRP-3 cells appeared higher in pre-ovulatory animals compared to all other stages. The percentage of GnRH cell bodies with kisspeptin appositions did not change with season or stage of estrous cycle. The percentage of kisspeptin cells receiving inputs from RFRP-3 fibers did not vary with season or stage of estrous cycle. These interactions suggest the possibility of the presence of an ultra-short loop feedback system between these three peptides. The changes in RFRP-3 neurons suggest the possibility of a role in the regulation of reproduction in the horse, but it is unlikely to be as a gonadotropin inhibitory factor.


Sujet(s)
Hormone de libération des gonadotrophines , Neuropeptides , Equus caballus , Animaux , Femelle , Hormone de libération des gonadotrophines/métabolisme , Kisspeptines/métabolisme , Saisons , Neuropeptides/physiologie , Hypothalamus/métabolisme , Cycle oestral/physiologie , Neurones
19.
Theriogenology ; 220: 108-115, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38507824

RÉSUMÉ

The presence of Kisspeptin (Kp) and its receptors in the corpus luteum (CL) of buffalo has recently been demonstrated. In this study, we investigated the role of Kp in the modulation of progesterone (P4) synthesis in vitro. The primary culture of bubaline luteal cells (LCs) was treated with 10, 50, and 100 nM of Kp and Kp antagonist (KpA) alongside a vehicle control. The combined effect of Kp and KpA was assessed at 100 nM concentration. Intracellular response to Kp treatment in the LCs was assessed by examining transcript profiles (LHR, STAR, CYP11A1, HSD3B1, and ERK1/2) using quantitative polymerase chain reaction (qPCR). In addition, the immunolocalization of ERK1/2 and phosphorylated ERK1/2 (p-ERK1/2) in the LCs was studied using immunocytochemistry. Accumulation of P4 from the culture supernatant was determined using enzyme-linked immunosorbent assay (ELISA). The results indicated that LCs had a greater p-ERK1/2 expression in the Kp treatment groups. A significant increase in the P4 concentration was recorded at 50 nM and 100 nM Kp, while KpA did not affect the basal concentration of P4. However, the addition of KpA to the Kp-treated group at 100 nM concentration suppressed the Kp-induced P4 accumulation into a concentration similar to the control. There was significant upregulation of ERK1/2 and CYP11A1 expressions in the Kp-treated LCs at 100 nM (18.1 and 37fold, respectively, p < 0.01). However, the addition of KpA to Kp-treated LCs modulated ERK1/2, LHR, STAR, CYP11A1, and HSD3B1 at 100 nM concentration. It can be concluded that Kp at 100 nM stimulated P4 production, while the addition of KpA suppressed Kp-induced P4 production in the buffalo LCs culture. Furthermore, an increment in p-ERK1/2 expression in the LCs indicated activation of the Kp signaling pathway was associated with luteal steroidogenesis.


Sujet(s)
Cellules lutéales , Femelle , Animaux , Progestérone/métabolisme , Kisspeptines/génétique , Kisspeptines/pharmacologie , Kisspeptines/métabolisme , Régulation positive , Extracellular Signal-Regulated MAP Kinases/métabolisme , Cholesterol side-chain cleavage enzyme/génétique , Cholesterol side-chain cleavage enzyme/métabolisme , Système de signalisation des MAP kinases , Corps jaune/physiologie , Complexes multienzymatiques/génétique , Complexes multienzymatiques/métabolisme
20.
Endocrinology ; 165(4)2024 Feb 20.
Article de Anglais | MEDLINE | ID: mdl-38470466

RÉSUMÉ

The neuroendocrine system that controls the preovulatory surge of gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH), which triggers ovulation in female mammals, is sexually differentiated in rodents. A transient increase in circulating testosterone levels in male rats within a few hours of birth is primarily responsible for the defeminization of anteroventral periventricular nucleus (AVPV) kisspeptin neurons, which are critical regulators of the GnRH/LH surge. The present study aimed to determine whether neonatal estradiol-17ß (E2) converted from testosterone by aromatase primarily causes the defeminization of AVPV kisspeptin neurons and the surge of GnRH/LH in male rodents. The results of the present study showed that the neonatal administration of letrozole (LET), a nonsteroidal aromatase inhibitor, within 2 hours of birth rescued AVPV Kiss1 expression and the LH surge in adult male rats, while the neonatal administration of testosterone propionate (TP) irreversibly attenuated AVPV Kiss1 expression and the LH surge in adult female rats. Furthermore, the neonatal LET-treated Kiss1-Cre-activated tdTomato reporter males exhibited a comparable number of AVPV Kiss1-Cre-activated tdTomato-expressing cells to that of vehicle-treated female rats, while neonatal TP-treated females showed fewer AVPV Kiss1-Cre-activated tdTomato-expressing cells than vehicle-treated females. Moreover, neonatal TP administration significantly decreased the number of arcuate Kiss1-expressing and Kiss1-Cre-activated tdTomato-positive cells and suppressed LH pulses in adult gonadectomized female rats; however, neonatal LET administration failed to affect them. These results suggest that E2 converted from neonatal testosterone is primarily responsible for the defeminization of AVPV kisspeptin neurons and the subsequent GnRH/LH surge generation in male rats.


Sujet(s)
Aromatase , Kisspeptines , , Animaux , Femelle , Mâle , Rats , Aromatase/métabolisme , Oestradiol/métabolisme , Hormone de libération des gonadotrophines/métabolisme , Hypothalamus antérieur/métabolisme , Kisspeptines/génétique , Kisspeptines/métabolisme , Hormone lutéinisante/métabolisme , Mammifères/métabolisme , Neurones/métabolisme , Testostérone/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...