Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
J Neuroendocrinol ; 34(10): e13201, 2022 10.
Article in English | MEDLINE | ID: mdl-36262016

ABSTRACT

The reproductive neuropeptide kisspeptin has emerged as the master regulator of mammalian reproduction due to its key roles in the initiation of puberty and the control of fertility. Alongside the tachykinin neurokinin B and the endogenous opioid dynorphin, these peptides are central to the hormonal control of reproduction. Building on the expanding body of experimental animal models, interest has flourished with human studies revealing that kisspeptin administration stimulates physiological reproductive hormone secretion in both healthy men and women, as well as patients with common reproductive disorders. In addition, emerging therapeutic roles based on neurokinin B for the management of menopausal flushing, endometriosis and uterine fibroids are increasingly recognised. In this review, we focus on kisspeptin and neurokinin B and their potential application as novel clinical strategies for the management of reproductive disorders.


Subject(s)
Kisspeptins , Neurokinin B , Male , Animals , Humans , Female , Neurokinin B/physiology , Kisspeptins/physiology , Reproductive Health , Dynorphins , Reproduction/physiology , Biology , Gonadotropin-Releasing Hormone , Mammals
2.
J Neuroendocrinol ; 34(5): e13119, 2022 05.
Article in English | MEDLINE | ID: mdl-35491543

ABSTRACT

In primates, the gonatotropin-releasing hormone (GnRH) neurosecretory system, consisting of GnRH, kisspeptin, and neurokinin B neurons, is active during the neonatal/early infantile period. During the late infantile period, however, activity of the GnRH neurosecretory system becomes minimal as a result of gonadal steroid independent central inhibition, and this suppressed GnRH neurosecretory state continues throughout the prepubertal period. At the initiation of puberty, the GnRH neurosecretory system becomes active again because of the decrease in central inhibition. During the progress of puberty, kisspeptin and neurokinin B signaling to GnRH neurons further increases, resulting in the release of gonadotropins and subsequent gonadal maturation, and hence puberty. This review further discusses potential substrates of central inhibition and subsequent pubertal modification of the GnRH neurosecretory system by the pubertal increase in steroid hormones, which ensures the regulation of adult reproductive function.


Subject(s)
Kisspeptins , Neurokinin B , Animals , Gonadotropin-Releasing Hormone , Kisspeptins/pharmacology , Luteinizing Hormone , Neurokinin B/physiology , Primates , Sexual Maturation/physiology
3.
J Pediatr Endocrinol Metab ; 34(3): 325-332, 2021 Mar 26.
Article in English | MEDLINE | ID: mdl-33675211

ABSTRACT

BACKGROUND: There is no data regarding the interrelationships of circulating Makorin Ring Finger Protein-3 (MKRN3), Kisspeptin (KISS1), and Neurokinin B (NKB) concentrations during minipuberty in humans. OBJECTIVE: To determine temporal changes in circulating concentrations of MKRN3, KISS1, NKB, and gonadotropins and investigate interrelationships between them in healthy full-term (FT) and preterm (PT) infants during minipuberty period. METHODS: A prospective study of 6-month follow-up performed. Eighty-seven healthy newborns, 48 FT (19 boys/29 girls), and 39 PT (21 boys/18 girls) (gestational age 31-37 weeks), were included. Blood samples were taken at 7 days (D7), 2 months (M2), and 6 months (M6) of age. Serum MKRN3, KISS1, NKB, LH, FSH, total testosterone (TT), and estradiol (E2) concentrations were measured. RESULTS: Seventy infants completed the study. MKRN3, KISS1, and NKB concentrations were similar in FT girls and boys. PT boys and girls also had similar concentrations of MKRN3, KISS1, and NKB. FT babies had significantly higher NKB concentrations than PT babies at D7, M2, and M6. MKRN3 and KISS1 concentrations do not differ between FT and PT babies. A strong positive correlation was found between MKRN3 and KISS1 at each time point and in all groups. FSH, LH, TT/E2 concentrations decrease while those of MKRN3 and KISS1 have a trend to increase toward the end of minipuberty. No correlation was detected between gonadotropins and MKRN3, KISS1, NKB concentrations. CONCLUSION: Strong positive correlation demonstrated between KISS1 and MKRN3 suggests that interrelationship between molecules controlling minipuberty is not similar to those at puberty.


Subject(s)
Hypothalamo-Hypophyseal System/physiology , Kisspeptins/physiology , Neurokinin B/physiology , Ovary/physiology , Testis/physiology , Ubiquitin-Protein Ligases/physiology , Female , Humans , Infant , Infant, Newborn , Luteinizing Hormone/blood , Male , Prospective Studies
4.
J Neuroendocrinol ; 33(3): e12945, 2021 03.
Article in English | MEDLINE | ID: mdl-33713519

ABSTRACT

Neurones in the arcuate nucleus co-expressing kisspeptin, neurokinin B (NKB) and dynorphin (KNDy) play a critical role in the control of gonadotrophin-releasing hormone (GnRH) and luteinising hormone (LH) secretion. In sheep, KNDy neurones mediate both steroid-negative- and -positive-feedback during pulsatile and preovulatory surge secretions of GnRH/LH, respectively. In addition, KNDy neurones receive glutamatergic inputs expressing vGlut2, a glutamate transporter that serves as a marker for those terminals, from both KNDy neurones and other populations of glutamatergic neurones. Previous work reported higher numbers of vGlut2-positive axonal inputs onto KNDy neurones during the LH surge than in luteal phase ewes. In the present study, we further examined the effects of the ovarian steroids progesterone (P) and oestradiol (E2 ) on glutamatergic inputs to KNDy neurones. Ovariectomised (OVX) ewes received either no further treatment (OVX) or steroid treatments that mimicked the luteal phase (low E2  + P), and early (low E2 ) or late follicular (high E2 ) phases of the oestrous cycle (n = 4 or 5 per group). Brain sections were processed for triple-label immunofluorescent detection of NKB/vGlut2/synaptophysin and analysed using confocal microscopy. We found higher numbers of vGlut2 inputs onto KNDy neurones in high E2 compared to the other three treatment groups. These results suggest that synaptic plasticity of glutamatergic inputs onto KNDy neurones during the ovine follicular phase depend on increasing levels of E2 required for the preovulatory GnRH/surge. These synaptic changes likely contribute to the positive-feedback action of oestrogen on GnRH/LH secretion and thus the generation of the preovulatory surge in the sheep.


Subject(s)
Dynorphins/physiology , Estradiol/physiology , Follicular Phase/physiology , Glutamates/physiology , Kisspeptins/physiology , Neurokinin B/physiology , Neuronal Plasticity/physiology , Synapses/physiology , Animals , Estradiol/metabolism , Female , Gonadotropin-Releasing Hormone/blood , Luteal Phase/drug effects , Luteinizing Hormone/blood , Ovariectomy , Sheep , Vesicular Glutamate Transport Protein 2/metabolism
5.
Domest Anim Endocrinol ; 74: 106522, 2021 01.
Article in English | MEDLINE | ID: mdl-32841888

ABSTRACT

Heat stress disrupts reproductive function in cattle. In summer, high ambient temperature and humidity elevate core body temperature, which is considered to be detrimental to reproductive abilities in cattle. Neurokinin B (NKB) is a factor that generates pulsatile GnRH and subsequent LH secretion in mammals. Recent studies have reported that NKB-neurokinin 3 receptor (NK3R) signaling is associated with heat-defense responses in rodents. The present study aimed to clarify the role of NKB-NK3R signaling in thermoregulation in cattle. We examined the effects of an NK3R-selective agonist, senktide, on vaginal temperature as an indicator of core body temperature in winter and summer. In both seasons, continuous infusion of senktide for 4 h immediately decreased vaginal temperature, and the mean temperature change in the senktide-treated group was significantly lower than that of both vehicle- and GnRH-treated groups. Administration of GnRH induced LH elevation, but there was no significant difference in vaginal temperature change between GnRH- and vehicle-treated groups. Moreover, we investigated the effects of senktide on ovarian temperature. Senktide treatment seemed to suppress the increase in ovarian temperature from 2 h after the beginning of administration, although the difference between groups was not statistically significant. Taken together, these results suggest that senktide infusion caused a decline in the vaginal temperature of cattle, in both winter and summer seasons, and this effect was not due to the gonadotropin-releasing action of senktide. These findings provide new therapeutic options for senktide to support both heat-defense responses and GnRH/LH pulse generation.


Subject(s)
Body Temperature/drug effects , Cattle/physiology , Heat-Shock Response/drug effects , Peptide Fragments/pharmacology , Receptors, Neurokinin-3/agonists , Substance P/analogs & derivatives , Animals , Body Temperature Regulation/drug effects , Body Temperature Regulation/physiology , Female , Gonadotropin-Releasing Hormone/physiology , Luteinizing Hormone/physiology , Neurokinin B/physiology , Ovary/physiology , Peptide Fragments/therapeutic use , Receptors, Neurokinin-3/physiology , Signal Transduction/physiology , Substance P/pharmacology , Substance P/therapeutic use , Vagina/physiology
6.
Nat Rev Endocrinol ; 16(8): 407-420, 2020 08.
Article in English | MEDLINE | ID: mdl-32427949

ABSTRACT

Hypothalamic kisspeptin neurons serve as the nodal regulatory centre of reproductive function. These neurons are subjected to a plethora of regulatory factors that ultimately affect the release of kisspeptin, which modulates gonadotropin-releasing hormone (GnRH) release from GnRH neurons to control the reproductive axis. The presence of sufficient energy reserves is critical to achieve successful reproduction. Consequently, metabolic factors impose a very tight control over kisspeptin synthesis and release. This Review offers a synoptic overview of the different steps in which kisspeptin neurons are subjected to metabolic regulation, from early developmental stages to adulthood. We cover an ample array of known mechanisms that underlie the metabolic regulation of KISS1 expression and kisspeptin release. Furthermore, the novel role of kisspeptin neurons as active players within the neuronal circuits that govern energy balance is discussed, offering evidence of a bidirectional role of these neurons as a nexus between metabolism and reproduction.


Subject(s)
Energy Metabolism/physiology , Kisspeptins/physiology , Reproduction/physiology , Animals , Dynorphins/physiology , Female , Gonadotropin-Releasing Hormone/physiology , Homeostasis , Humans , Hypothalamo-Hypophyseal System/physiology , Hypothalamus/cytology , Hypothalamus/physiology , Kisspeptins/genetics , Luteinizing Hormone/physiology , Neurokinin B/physiology , Neurons/physiology , Ovary/physiology , Puberty/physiology
7.
Sci China Life Sci ; 63(6): 875-885, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32266609

ABSTRACT

Brain-to-brain interfaces (BtBIs) hold exciting potentials for direct communication between individual brains. However, technical challenges often limit their performance in rapid information transfer. Here, we demonstrate an optical brain-to-brain interface that transmits information regarding locomotor speed from one mouse to another and allows precise, real-time control of locomotion across animals with high information transfer rate. We found that the activity of the genetically identified neuromedin B (NMB) neurons within the nucleus incertus (NI) precisely predicts and critically controls locomotor speed. By optically recording Ca2+ signals from the NI of a "Master" mouse and converting them to patterned optogenetic stimulations of the NI of an "Avatar" mouse, the BtBI directed the Avatar mice to closely mimic the locomotion of their Masters with information transfer rate about two orders of magnitude higher than previous BtBIs. These results thus provide proof-of-concept that optical BtBIs can rapidly transmit neural information and control dynamic behaviors across individuals.


Subject(s)
Brain-Computer Interfaces , Brain/physiology , Locomotion/physiology , Optical Imaging/methods , Animals , Behavior Control , Behavior, Animal/physiology , Calcium/metabolism , Calcium Signaling/physiology , Computer Simulation , Dependovirus/metabolism , HEK293 Cells , Humans , Kinetics , Mice , Models, Biological , Neurokinin B/analogs & derivatives , Neurokinin B/physiology , Neurons/physiology , Raphe Nuclei/physiology , Support Vector Machine , Transfection
8.
Gen Comp Endocrinol ; 287: 113342, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31783025

ABSTRACT

Copper is a metal ion present in all organisms, where it has well-known roles in association with proteins and enzymes essential for cellular processes. In the early decades of the twentieth century copper was shown to influence mammalian reproductive biology, and it was subsequently shown to exert effects primarily at the level of the pituitary gland and/or hypothalamic regions of the brain. Furthermore, it has been reported that copper can interact with key neuropeptides in the hypothalamic-pituitary-gonadal axis, notably gonadotropin-releasing hormone (GnRH) and neurokinin B. Interestingly, recent phylogenetic analysis of the sequences of GnRH-related peptides indicates that copper binding is an evolutionarily ancient property of this neuropeptide family, which has been variously retained, modified or lost in the different taxa. In this mini-review the metal-binding properties of neuropeptides in the vertebrate reproductive pathway are reviewed and the evolutionary and functional significance of copper binding by GnRH-related neuropeptides in vertebrates and invertebrates are discussed.


Subject(s)
Copper/pharmacology , Endocrine System/drug effects , Gonadotropin-Releasing Hormone/drug effects , Neurokinin B/drug effects , Reproduction/drug effects , Animals , Endocrine System/physiology , Gonadotropin-Releasing Hormone/chemistry , Gonadotropin-Releasing Hormone/physiology , Invertebrates/metabolism , Mammals/metabolism , Neurokinin B/chemistry , Neurokinin B/physiology , Protein Conformation/drug effects , Reproduction/physiology , Structure-Activity Relationship , Vertebrates/metabolism
10.
Semin Reprod Med ; 37(3): 119-124, 2019 05.
Article in English | MEDLINE | ID: mdl-31869839

ABSTRACT

Kisspeptin and neurokinin B (NKB) are hypothalamic neuropeptides that are vital for reproductive health. An absence of either kisspeptin or NKB signaling results in hypogonadotrophic hypogonadism and a failure to proceed through puberty. In recent years, several studies have demonstrated potential avenues for the clinical utility of medications that act through these pathways in the assessment and treatment of reproductive disorders. Kisspeptin acts to stimulate hypothalamic gonadotrophic-releasing hormone (GnRH) secretion from the hypothalamus. Kisspeptin induces gonadotrophin secretion in both healthy men and women, and in women with reproductive disorders such as hypothalamic amenorrhea (HA). Kisspeptin-based treatments hold promise for use during in vitro fertilization (IVF) treatment; a bolus of kisspeptin-54 induces an LH surge of 12 to 14 hours of duration sufficient to induce oocyte maturation, but with markedly reduced rates of the most significant complication of IVF treatment, ovarian hyperstimulation syndrome (OHSS). Kisspeptin could also be used chronically to restore reproductive health in patients with functional hypogonadism, such as those with HA. Furthermore, kisspeptin has potential as a diagnostic test of hypothalamic function; a "kisspeptin test" could be used in children with delayed puberty to identify the subset with genetically determined deficits in hypothalamic pathways (congenital hypogonadotrophic hypogonadism [CHH]). In addition to its role in hypothalamic GnRH pulse generation, NKB plays a critical role in the occurrence of one of the most troubling symptoms of the menopause, the "hot flush." Neurokinin-3 receptor (NK3R) antagonists are highly effective as treatments for hot flushes in postmenopausal women, with several compounds now in late-phase development. Furthermore, NK3R antagonism leads to a reduction in LH secretion by reducing GnRH pulsatility in the hypothalamus and has been shown to reduce androgen levels in women with polycystic ovary syndrome (PCOS) (in whom GnRH pulsatility is often increased). In summary, although further detailed evaluation in several clinical settings is ongoing, medications based on kisspeptin and NKB pathways have prodigious potential in the assessment and treatment of reproductive disorders.


Subject(s)
Kisspeptins/physiology , Neurokinin B/physiology , Translational Research, Biomedical , Animals , Female , Fertilization in Vitro/methods , Fertilization in Vitro/trends , Gonadotropin-Releasing Hormone/metabolism , Humans , Hypothalamus/metabolism , Male , Reproduction/physiology , Translational Research, Biomedical/methods , Translational Research, Biomedical/trends
11.
Semin Reprod Med ; 37(3): 125-130, 2019 05.
Article in English | MEDLINE | ID: mdl-31869840

ABSTRACT

Vasomotor symptoms, including hot flushes and night sweats, pose a significant symptomatic burden to women undergoing menopause, and negatively impact on both their physical and psychological well-being. Management of these symptoms can be challenging, with the use of conventional hormone replacement therapy limited by duration of treatment and clinical contraindications. Recent advances in our understanding of the neuroendocrine regulation of the reproductive axis and thermoregulation postmenopause has helped identify a promising new therapeutic target to ameliorate hot flushes. Antagonism of the neurokinin B/neurokinin-3 receptor (NK3R) signaling pathway has emerged as an efficacious treatment in managing vasomotor symptoms, with evidence of rapid and sustained reduction in hot flush frequency and severity and improvements in secondary quality-of-life measures such as sleep. Within this review, we will explore the growing body of evidence supporting the use of NK3R antagonists in the management of vasomotor symptoms, and the possible utility in managing dysfunctional sex-hormone-dependent disorders and glycolipid metabolism disorders such as polycystic ovarian syndrome.


Subject(s)
Hot Flashes/etiology , Hot Flashes/therapy , Menopause/physiology , Neurokinin B/physiology , Receptors, Neurokinin-3/physiology , Female , Hormone Replacement Therapy/methods , Humans , Quality of Life , Signal Transduction/physiology , Vasomotor System/physiology , Vasomotor System/physiopathology
12.
J Neurosci ; 39(49): 9738-9747, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31645462

ABSTRACT

Fertility critically depends on the gonadotropin-releasing hormone (GnRH) pulse generator, a neural construct comprised of hypothalamic neurons coexpressing kisspeptin, neurokoinin-B and dynorphin. Here, using mathematical modeling and in vivo optogenetics we reveal for the first time how this neural construct initiates and sustains the appropriate ultradian frequency essential for reproduction. Prompted by mathematical modeling, we show experimentally using female estrous mice that robust pulsatile release of luteinizing hormone, a proxy for GnRH, emerges abruptly as we increase the basal activity of the neuronal network using continuous low-frequency optogenetic stimulation. Further increase in basal activity markedly increases pulse frequency and eventually leads to pulse termination. Additional model predictions that pulsatile dynamics emerge from nonlinear positive and negative feedback interactions mediated through neurokinin-B and dynorphin signaling respectively are confirmed neuropharmacologically. Our results shed light on the long-elusive GnRH pulse generator offering new horizons for reproductive health and wellbeing.SIGNIFICANCE STATEMENT The gonadotropin-releasing hormone (GnRH) pulse generator controls the pulsatile secretion of the gonadotropic hormones LH and FSH and is critical for fertility. The hypothalamic arcuate kisspeptin neurons are thought to represent the GnRH pulse generator, since their oscillatory activity is coincident with LH pulses in the blood; a proxy for GnRH pulses. However, the mechanisms underlying GnRH pulse generation remain elusive. We developed a mathematical model of the kisspeptin neuronal network and confirmed its predictions experimentally, showing how LH secretion is frequency-modulated as we increase the basal activity of the arcuate kisspeptin neurons in vivo using continuous optogenetic stimulation. Our model provides a quantitative framework for understanding the reproductive neuroendocrine system and opens new horizons for fertility regulation.


Subject(s)
Gonadotropin-Releasing Hormone/physiology , Animals , Dynorphins/physiology , Estrous Cycle/physiology , Feedback, Physiological , Female , Kisspeptins/physiology , Luteinizing Hormone/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Theoretical , Neurokinin B/physiology , Neurons/physiology , Optogenetics , Pregnancy , Reproduction/physiology , Ultradian Rhythm/physiology
13.
F1000Res ; 82019.
Article in English | MEDLINE | ID: mdl-31297186

ABSTRACT

This review recounts the origins and development of the concept of the hypothalamic gonadotropin-releasing hormone (GnRH) pulse generator. It starts in the late 1960s when striking rhythmic episodes of luteinizing hormone secretion, as reflected by circulating concentrations of this gonadotropin, were first observed in monkeys and ends in the present day. It is currently an exciting time witnessing the application, primarily to the mouse, of contemporary neurobiological approaches to delineate the mechanisms whereby Kiss1/NKB/Dyn (KNDy) neurons in the arcuate nucleus of the hypothalamus generate and time the pulsatile output of kisspeptin from their terminals in the median eminence that in turn dictates intermittent GnRH release and entry of this decapeptide into the primary plexus of the hypophysial portal circulation. The review concludes with an examination of questions that remain to be addressed.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiology , Gonadotropin-Releasing Hormone/physiology , Kisspeptins/physiology , Animals , Dynorphins/physiology , Mice , Neurokinin B/physiology , Neurons/physiology
15.
Reproduction ; 156(3): R83-R99, 2018 09.
Article in English | MEDLINE | ID: mdl-29880718

ABSTRACT

Early work in ewes provided a wealth of information on the physiological regulation of pulsatile gonadotropin-releasing hormone (GnRH) secretion by internal and external inputs. Identification of the neural systems involved, however, was limited by the lack of information on neural mechanisms underlying generation of GnRH pulses. Over the last decade, considerable evidence supported the hypothesis that a group of neurons in the arcuate nucleus that contain kisspeptin, neurokinin B and dynorphin (KNDy neurons) are responsible for synchronizing secretion of GnRH during each pulse in ewes. In this review, we describe our current understanding of the neural systems mediating the actions of ovarian steroids and three external inputs on GnRH pulsatility in light of the hypothesis that KNDy neurons play a key role in GnRH pulse generation. In breeding season adults, estradiol (E2) and progesterone decrease GnRH pulse amplitude and frequency, respectively, by actions on KNDy neurons, with E2 decreasing kisspeptin and progesterone increasing dynorphin release onto GnRH neurons. In pre-pubertal lambs, E2 inhibits GnRH pulse frequency by decreasing kisspeptin and increasing dynorphin release, actions that wane as the lamb matures to allow increased pulsatile GnRH secretion at puberty. Less is known about mediators of undernutrition and stress, although some evidence implicates kisspeptin and dynorphin, respectively, in the inhibition of GnRH pulse frequency by these factors. During the anoestrus, inhibitory photoperiod acting via melatonin activates A15 dopaminergic neurons that innervate KNDy neurons; E2 increases dopamine release from these neurons to inhibit KNDy neurons and suppress the frequency of kisspeptin and GnRH release.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Homeostasis/physiology , Sheep/physiology , Animals , Arcuate Nucleus of Hypothalamus/physiology , Breeding , Dynorphins/physiology , Estradiol/pharmacology , Estrous Cycle , Feedback, Physiological , Female , Kisspeptins/physiology , Luteinizing Hormone/metabolism , Neurokinin B/physiology , Neurons/physiology , Periodicity , Progesterone/pharmacology , Seasons , Sexual Maturation/physiology
16.
Gynecol Endocrinol ; 34(11): 913-919, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29902942

ABSTRACT

Women during perimenopausal period experience a range of symptoms, which interfere with physical, sexual, and social life. About 65-75% of symptoms connected with postmenopausal period are vasomotor symptoms (VMS), such as hot flushes and night sweats. Hot flushes are subjective sensation of heat associated with cutaneous vasodilatation and drop in core temperature. It is suspected that VMS are strongly correlated with pulsatile oversecretion of gonadotropin-releasing hormone (GnRH) and subsequently luteinizing hormone (LH). Evidence has accumulated in parallel showing that lack of negative feedback of steroid hormones synthesized in ovary causes overactivation of hypertrophied kisspeptin/neurokinin B/dynorphin (KNDy) neurons, located in infundibular nucleus. Oversecretion of both kisspeptin (KISS1) and neurokinin B (NKB), as well as downregulation of dynorphin, plays dominant role in creation of GnRH pulses. This in turn causes VMS. Administration of senktide, highly potent and selective NK3R agonist, resulted in increase of serum LH concentration, induction of VMS, increase in heart rate, and skin temperature in postmenopausal women. These finding suggest that modulation of KNDy neurons may become new therapeutic approach in the treatment of VMS.


Subject(s)
Hot Flashes/etiology , Hypothalamus/physiology , Neurons/physiology , Postmenopause/physiology , Vasomotor System/physiology , Dynorphins/physiology , Feedback, Physiological , Female , Hot Flashes/drug therapy , Humans , Kisspeptins/physiology , Neurokinin B/physiology
17.
Curr Opin Endocrinol Diabetes Obes ; 25(1): 36-41, 2018 02.
Article in English | MEDLINE | ID: mdl-29120926

ABSTRACT

PURPOSE OF REVIEW: The current review highlights recent advances in physiological and pharmacological researches in biology of mammalian bombesin-like peptides (BLPs). RECENT FINDINGS: BLPs and their receptors were found to have regulatory roles in many biological processes in central nervous system. Two BLPs, neuromedin B and gastrin-releasing peptide (GRP), and their receptors are required for regulation of basal and induced sighing activity in rodents. This is the first study demonstrating central pathways involved in regulation of sighing activity. GRP receptor (GRPR) expressing neurons are excitatory glutamatergic interneurons located in the dorsal lamina without projections outside the spinal cord and mediate itch signals via vesicular glutamate transporter 2. Those neurons receive itch signals and make synapses with the parabrachial nucleus projecting spinal neurons to transmit itch signals to parabrachial nucleus. GRP expressing interneurons function in a proposed 'leaky gate model' to interpret the mechanism of both pain and itch transmission. In addition to recent advances of biology in nervous system, BLPs and their receptors were found to play potential regulatory roles in innate and adaptive immune responses and tissue development. SUMMARY: Several important biological roles of BLPs and their receptors in nervous system were identified. Together with researches regarding central roles of BLPs, studies revealing the regulatory roles of BLPs and their receptors in immunology and tissue development provide us with novel insights into understanding of the biology of BLPs and their receptors.


Subject(s)
Biological Phenomena , Gastrin-Releasing Peptide/physiology , Neurokinin B/analogs & derivatives , Receptors, Bombesin/physiology , Animals , Bombesin/chemistry , Bombesin/metabolism , Gastrin-Releasing Peptide/chemistry , Glucose/metabolism , Humans , Neurokinin B/chemistry , Neurokinin B/physiology , Organogenesis/genetics , Pain Perception/physiology , Pruritus/genetics , Pruritus/metabolism
18.
Endocrinology ; 158(10): 3269-3280, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28977601

ABSTRACT

Loss-of-function or inactivating mutations in the genes coding for kisspeptin and its receptor (KISS1R) or neurokinin B (NKB) and the NKB receptor (NK3R) in humans result in a delay in or the absence of puberty. However, precise mechanisms of kisspeptin and NKB signaling in the regulation of the pubertal increase in gonadotropin-releasing hormone (GnRH) release in primates are unknown. In this study, we conducted a series of experiments infusing agonists and antagonists of kisspeptin and NKB into the stalk-median eminence, where GnRH, kisspeptin, and NKB neuroterminal fibers are concentrated, and measuring GnRH release in prepubertal and pubertal female rhesus monkeys. Results indicate that (1) similar to those previously reported for GnRH stimulation by the KISS1R agonist (i.e., human kisspeptin-10), the NK3R agonist senktide stimulated GnRH release in a dose-responsive manner in both prepubertal and pubertal monkeys; (2) the senktide-induced GnRH release was blocked in the presence of the KISS1R antagonist peptide 234 in pubertal but not prepubertal monkeys; and (3) the kisspeptin-induced GnRH release was blocked in the presence of the NK3R antagonist SB222200 in the pubertal but not prepubertal monkeys. These results are interpreted to mean that although, in prepubertal female monkeys, kisspeptin and NKB signaling to GnRH release is independent, in pubertal female monkeys, a reciprocal signaling mechanism between kisspeptin and NKB neurons is established. We speculate that this cooperative mechanism by the kisspeptin and NKB network underlies the pubertal increase in GnRH release in female monkeys.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Kisspeptins/physiology , Macaca mulatta/physiology , Neurokinin B/physiology , Sexual Maturation/physiology , Signal Transduction/physiology , Animals , Female , Kisspeptins/agonists , Kisspeptins/antagonists & inhibitors , Kisspeptins/pharmacology , Median Eminence/drug effects , Neurokinin B/agonists , Neurokinin B/antagonists & inhibitors , Neurons/metabolism , Peptide Fragments/pharmacology , Quinolines/pharmacology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, Kisspeptin-1 , Receptors, Neurokinin-3/agonists , Signal Transduction/drug effects , Substance P/analogs & derivatives , Substance P/pharmacology
19.
J Neuroendocrinol ; 29(6)2017 06.
Article in English | MEDLINE | ID: mdl-28475285

ABSTRACT

Pulsatile secretion of gonadotrophin-releasing hormone (GnRH)/luteinising hormone is indispensable for the onset of puberty and reproductive activities at adulthood in mammalian species. A cohort of neurones expressing three neuropeptides, namely kisspeptin, encoded by the Kiss1 gene, neurokinin B (NKB) and dynorphin A, localised in the hypothalamic arcuate nucleus (ARC), so-called KNDy neurones, comprises a putative intrinsic source of the GnRH pulse generator. Synchronous activity among KNDy neurones is considered to be required for pulsatile GnRH secretion. It has been reported that gap junctions play a key role in synchronising electrical activity in the central nervous system. Thus, we hypothesised that gap junctions are involved in the synchronised activities of KNDy neurones, which is induced by NKB-NK3R signalling. We determined the role of NKB-NK3R signalling in Ca2+ oscillation (an indicator of neuronal activities) of KNDy neurones and its synchronisation mechanism among KNDy neurones. Senktide, a selective agonist for NK3R, increased the frequency of Ca2+ oscillations in cultured Kiss1-GFP cells collected from the mediobasal hypothalamus of the foetal Kiss1-green fluorescent protein (GFP) mice. The senktide-induced Ca2+ oscillations were synchronised in the Kiss1-GFP and neighbouring glial cells. Confocal microscopy analysis of these cells, which have shown synchronised Ca2+ oscillations, revealed close contacts between Kiss1-GFP cells, as well as between Kiss1-GFP cells and glial cells. Dye coupling experiments suggest cell-to-cell communication through gap junctions between Kiss1-GFP cells and neighbouring glial cells. Connexin-26 and -37 mRNA were found in isolated ARC Kiss1 cells taken from adult female Kiss1-GFP transgenic mice. Furthermore, 18ß-glycyrrhetinic acids and mefloquine, which are gap junction inhibitors, attenuated senktide-induced Ca2+ oscillations in Kiss1-GFP cells. Taken together, these results suggest that NKB-NK3R signalling enhances synchronised activities among neighbouring KNDy neurones, and that both neurone-neurone and neurone-glia communications via gap junctions possibly contribute to synchronised activities among KNDy neurones.


Subject(s)
Gap Junctions/physiology , Neuroglia/physiology , Neurons/physiology , Peptide Fragments/pharmacology , Substance P/analogs & derivatives , Animals , Cells, Cultured , Connexins/metabolism , Dynorphins/physiology , Gap Junctions/drug effects , Gap Junctions/metabolism , Glycyrrhetinic Acid/analogs & derivatives , Glycyrrhetinic Acid/pharmacology , Kisspeptins/genetics , Medulla Oblongata/metabolism , Mefloquine/pharmacology , Mice, Transgenic , Neuroglia/metabolism , Neurokinin B/physiology , Neurons/drug effects , Neurons/metabolism , Peptide Fragments/antagonists & inhibitors , Substance P/antagonists & inhibitors , Substance P/pharmacology
20.
J Endocrinol ; 233(2): 159-174, 2017 05.
Article in English | MEDLINE | ID: mdl-28330973

ABSTRACT

Kisspeptin and neurokinin B (NKB) are neuropeptides co-expressed in the mammalian hypothalamus and coordinately control GnRH signaling. We have found that Nkb and kisspeptin neurons are distinct in the teleost, striped bass (STB) and capitalized on this phenomenon to study the mode of action of Nkb and its related neuropeptide-F (Nkf), both of which are encoded by the tac3 gene. In vitro brain slices and in vivo administration studies revealed that Nkb/f consistently downregulated kiss2, whereas antagonist (AntD) administration restored this effect. Overall, a minor effect was noted on gnrh1 expression, whereas Gnrh1 content in the pituitaries was reduced after Nkb/f treatment and increased with AntD. Concomitantly, immunostaining demonstrated that hypothalamic Nkb neurons border and densely innervate the largest kiss2 neuronal population in the hypothalamus, which also coexpresses Nkb receptor. No expression of Nkb receptor or Nkb neuronal projections was detected near/in Gnrh1 soma in the preoptic area. At the level of the pituitary, however, the picture was more complex: both Nkb/f and AntD upregulated lhb and fshb expression and Lh secretion in vivo Together with the stimulatory effect of Nkb/f on Lh/Fsh secretion from pituitary cells, in vitro, this may indicate an additional independent action of Nkb/f within the pituitary, in which the hypothalamic pathway is more dominant. The current study demonstrates that Nkb/f utilizes multiple pathways to regulate reproduction in the STB and that in the brain, Nkb mainly acts as a negative modulator of kiss2 to regulate the release of Gnrh1.


Subject(s)
Bass/metabolism , Gene Expression Regulation/physiology , Kisspeptins/metabolism , Neurokinin B/physiology , Reproduction/physiology , Animals , Cloning, Molecular , DNA, Complementary/metabolism , Kisspeptins/antagonists & inhibitors , Kisspeptins/genetics , Male , Neurokinin B/genetics , Pituitary Gland/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...