Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
Add more filters










Publication year range
1.
Endocrinology ; 164(11)2023 09 23.
Article in English | MEDLINE | ID: mdl-37776515

ABSTRACT

The current model for the synchronization of GnRH neural activity driving GnRH and LH pulses proposes that a set of arcuate (ARC) neurons that contain kisspeptin, neurokinin B, and dynorphin (KNDy neurons) is the GnRH pulse generator. This study tested the functional role of ovine KNDy neurons in pulse generation and explored the roles of nearby Kiss1 receptor (Kiss1R)-containing cells using lesions produced with saporin (SAP) conjugates. Injection of NK3-SAP ablated over 90% of the KNDy cells, while Kiss-SAP (saporin conjugated to kisspeptin-54) lesioned about two-thirds of the Kiss1R population without affecting KNDy or GnRH cell number. Both lesions produced a dramatic decrease in LH pulse amplitude but had different effects on LH pulse patterns. NK3-SAP increased interpulse interval, but Kiss-SAP did not. In contrast, Kiss-SAP disrupted the regular hourly occurrence of LH pulses, but NK3-SAP did not. Because Kiss1R is not expressed in KNDy cells, HiPlex RNAScope was used to assess the colocalization of 8 neurotransmitters and 3 receptors in ARC Kiss1R-containing cells. Kiss1R cells primarily contained transcript markers for GABA (68%), glutamate (28%), ESR1 (estrogen receptor-α) mRNA, and OPRK1 (kappa opioid receptor) mRNA. These data support the conclusion that KNDy neurons are essential for GnRH pulses in ewes, whereas ARC Kiss1R cells are not but do maintain the amplitude and regularity of GnRH pulses. We thus propose that in sheep, ARC Kiss1R neurons form part of a positive feedback circuit that reinforces the activity of the KNDy neural network, with GABA or glutamate likely being involved.


Subject(s)
Arcuate Nucleus of Hypothalamus , Kisspeptins , Luteinizing Hormone , Neurons , Animals , Female , Arcuate Nucleus of Hypothalamus/metabolism , Dynorphins/metabolism , gamma-Aminobutyric Acid , Glutamates , Gonadotropin-Releasing Hormone/metabolism , Kisspeptins/metabolism , Neurokinin B/metabolism , Neurons/metabolism , Receptors, Kisspeptin-1/genetics , RNA, Messenger , Saporins , Sheep , Luteinizing Hormone/metabolism
2.
Front Endocrinol (Lausanne) ; 13: 951344, 2022.
Article in English | MEDLINE | ID: mdl-35992143

ABSTRACT

Polycystic ovary syndrome (PCOS) is associated with elevated androgen and luteinizing hormone (LH) secretion and with oligo/anovulation. Evidence indicates that elevated androgens impair sex steroid hormone feedback regulation of pulsatile LH secretion. Hyperandrogenemia in PCOS may also disrupt the preovulatory LH surge. The mechanisms through which this might occur, however, are not fully understood. Kisspeptin (KISS1) neurons of the rostral periventricular area of the third ventricle (RP3V) convey hormonal cues to gonadotropin-releasing hormone (GnRH) neurons. In rodents, the preovulatory surge is triggered by these hormonal cues and coincident timing signals from the central circadian clock in the suprachiasmatic nucleus (SCN). Timing signals are relayed to GnRH neurons, in part, via projections from SCN arginine-vasopressin (AVP) neurons to RP3VKISS1 neurons. Because rodent SCN cells express androgen receptors (AR), we hypothesized that these circuits are impaired by elevated androgens in a mouse model of PCOS. In prenatally androgen-treated (PNA) female mice, SCN Ar expression was significantly increased compared to that found in prenatally vehicle-treated mice. A similar trend was seen in the number of Avp-positive SCN cells expressing Ar. In the RP3V, the number of kisspeptin neurons was preserved. Anterograde tract-tracing, however, revealed reduced SCNAVP neuron projections to the RP3V and a significantly lower proportion of RP3VKISS1 neurons with close appositions from SCNAVP fibers. Functional assessments showed, on the other hand, that RP3VKISS1 neuron responses to AVP were maintained in PNA mice. These findings indicate that PNA changes some of the neural circuits that regulate the preovulatory surge. These impairments might contribute to ovulatory dysfunction in PNA mice modeling PCOS.


Subject(s)
Kisspeptins , Polycystic Ovary Syndrome , Suprachiasmatic Nucleus , Androgens/metabolism , Androgens/pharmacology , Animals , Arginine , Arginine Vasopressin/metabolism , Female , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Humans , Kisspeptins/genetics , Kisspeptins/metabolism , Luteinizing Hormone/metabolism , Mice , Neurons/metabolism , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , Pregnancy , Suprachiasmatic Nucleus/metabolism , Vasopressins/metabolism
3.
J Neuroendocrinol ; 34(5): e13115, 2022 05.
Article in English | MEDLINE | ID: mdl-35502534

ABSTRACT

The anatomy and morphology of gonadotropin-releasing hormone (GnRH) neurons makes them both a joy and a challenge to investigate. They are a highly unique population of neurons given their developmental migration into the brain from the olfactory placode, their relatively small number, their largely scattered distribution within the rostral forebrain, and, in some species, their highly varied individual anatomical characteristics. These unique features have posed technological hurdles to overcome and promoted fertile ground for the establishment and use of creative approaches. Historical and more contemporary discoveries defining GnRH neuron anatomy remain critical in shaping and challenging our views of GnRH neuron function in the regulation of reproductive function. We begin this review with a historical overview of anatomical discoveries and developing methodologies that have shaped our understanding of the reproductive axis. We then highlight significant discoveries across specific groups of mammalian species to address some of the important comparative aspects of GnRH neuroanatomy. Lastly, we touch on unresolved questions and opportunities for future neuroanatomical research on this fascinating and important population of neurons.


Subject(s)
Gonadotropin-Releasing Hormone , Neuroanatomy , Animals , Gonadotropin-Releasing Hormone/metabolism , Mammals , Neurons/metabolism , Prosencephalon , Reproduction
4.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Article in English | MEDLINE | ID: mdl-35110409

ABSTRACT

A hypothalamic pulse generator located in the arcuate nucleus controls episodic release of gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH) and is essential for reproduction. Recent evidence suggests this generator is composed of arcuate "KNDy" cells, the abbreviation based on coexpression of kisspeptin, neurokinin B, and dynorphin. However, direct visual evidence of KNDy neuron activity at a single-cell level during a pulse is lacking. Here, we use in vivo calcium imaging in freely moving female mice to show that individual KNDy neurons are synchronously activated in an episodic manner, and these synchronized episodes always precede LH pulses. Furthermore, synchronization among KNDy cells occurs in a temporal order, with some subsets of KNDy cells serving as "leaders" and others as "followers" during each synchronized episode. These results reveal an unsuspected temporal organization of activation and synchronization within the GnRH pulse generator, suggesting that different subsets of KNDy neurons are activated at pulse onset than afterward during maintenance and eventual termination of each pulse. Further studies to distinguish KNDy "leader" from "follower" cells is likely to have important clinical significance, since regulation of pulsatile GnRH secretion is essential for normal reproduction and disrupted in pathological conditions such as polycystic ovary syndrome and hypothalamic amenorrhea.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Neurons/metabolism , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Dynorphins/metabolism , Female , Kisspeptins/metabolism , Luteinizing Hormone/metabolism , Mice , Mice, Inbred C57BL , Neurokinin B/metabolism , Reproduction/physiology
5.
Endocrinology ; 162(11)2021 11 01.
Article in English | MEDLINE | ID: mdl-34346492

ABSTRACT

Polycystic ovarian syndrome (PCOS), the most common endocrinopathy affecting women worldwide, is characterized by elevated luteinizing hormone (LH) pulse frequency due to the impaired suppression of gonadotrophin-releasing hormone (GnRH) release by steroid hormone negative feedback. Although neurons that co-express kisspeptin, neurokinin B, and dynorphin (KNDy cells) were recently defined as the GnRH/LH pulse generator, little is understood about their role in the pathogenesis of PCOS. We used a prenatal androgen-treated (PNA) mouse model of PCOS to determine whether changes in KNDy neurons or their afferent network underlie altered negative feedback. First, we identified elevated androgen receptor gene expression in KNDy cells of PNA mice, whereas progesterone receptor and dynorphin gene expression was significantly reduced, suggesting elevated androgens in PCOS disrupt progesterone negative feedback via direct actions upon KNDy cells. Second, we discovered GABAergic and glutamatergic synaptic input to KNDy neurons was reduced in PNA mice. Retrograde monosynaptic tract-tracing revealed a dramatic reduction in input originates from sexually dimorphic afferents in the preoptic area, anteroventral periventricular nucleus, anterior hypothalamic area and lateral hypothalamus. These results reveal 2 sites of neuronal alterations potentially responsible for defects in negative feedback in PCOS: changes in gene expression within KNDy neurons, and changes in synaptic inputs from steroid hormone-responsive hypothalamic regions. How each of these changes contribute to the neuroendocrine phenotype seen in in PCOS, and the role of specific sets of upstream KNDy afferents in the process, remains to be determined.


Subject(s)
Androgens/blood , Neurons/pathology , Polycystic Ovary Syndrome/pathology , Prenatal Exposure Delayed Effects , Afferent Pathways/drug effects , Afferent Pathways/metabolism , Androgens/pharmacology , Animals , Disease Models, Animal , Dynorphins/metabolism , Female , Kisspeptins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurokinin B/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Neurons, Afferent/drug effects , Neurons, Afferent/metabolism , Neurosecretory Systems/drug effects , Neurosecretory Systems/metabolism , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/physiopathology , Polycystic Ovary Syndrome/psychology , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/psychology
6.
Front Neurol ; 12: 670536, 2021.
Article in English | MEDLINE | ID: mdl-34239493

ABSTRACT

Spinal cord injury (SCI) in men is commonly associated with sexual dysfunction, including anejaculation, and chronic mid-thoracic contusion injury in male rats also impairs ejaculatory reflexes. Ejaculation is controlled by a spinal ejaculation generator consisting of a population of lumbar spinothalamic (LSt) neurons that control ejaculation through release of four neuropeptides including galanin and gastrin releasing peptide (GRP) onto lumbar and sacral autonomic and motor nuclei. It was recently demonstrated that spinal contusion injury in male rats caused reduction of GRP-immunoreactivity, but not galanin-immunoreactivity in LSt cells, indicative of reduced GRP peptide levels, but inconclusive results for galanin. The current study further tests the hypothesis that contusion injury causes a disruption of GRP and galanin mRNA in LSt cells. Male rats received mid-thoracic contusion injury and galanin and GRP mRNA were visualized 8 weeks later in the lumbar spinal cord using fluorescent in situ hybridization. Spinal cord injury significantly reduced GRP and galanin mRNA in LSt cells. Galanin expression was higher in LSt cells compared to GRP. However, expression of the two transcripts were positively correlated in LSt cells in both sham and SCI animals, suggesting that expression for the two neuropeptides may be co-regulated. Immunofluorescent visualization of galanin and GRP peptides demonstrated a significant reduction in GRP-immunoreactivity, but not galanin in LSt cells, confirming the previous observations. In conclusion, SCI reduced GRP and galanin expression in LSt cells with an apparent greater impact on GRP peptide levels. GRP and galanin are both essential for triggering ejaculation and thus such reduction may contribute to ejaculatory dysfunction following SCI in rats.

7.
Front Neurol ; 11: 983, 2020.
Article in English | MEDLINE | ID: mdl-33013649

ABSTRACT

20-HETE is a potent vasoconstrictor that is implicated in the regulation of blood pressure, cerebral blood flow and neuronal death following ischemia. Numerous human genetic studies have shown that inactivating variants in the cytochrome P450 enzymes that produce 20-HETE are associated with hypertension, stroke and cerebrovascular disease. However, little is known about the expression and cellular distribution of the cytochrome P450A enzymes (CYP4A) that produce 20-HETE or the newly discovered 20-HETE receptor (GPR75) in the brain. The present study examined the cell types and regions in the rat forebrain that express CYP4A and GPR75. Brain tissue slices from Sprague Dawley (SD), Dahl Salt-Sensitive (SS) and CYP4A1 transgenic rat strains, as well as cultured human cerebral pericytes and cerebral vascular smooth muscle cells, were analyzed by fluorescent immunostaining. Tissue homogenates from these strains and cultured cells were examined by Western blot. In the cerebral vasculature, CYP4A and GPR75 were expressed in endothelial cells, vascular smooth muscle cells and the glial limiting membrane of pial arteries and penetrating arterioles but not in the endothelium of capillaries. CYP4A, but not GPR75, was expressed in astrocytes. CYP4A and GPR75 were both expressed in a subpopulation of pericytes on capillaries. The diameters of capillaries were significantly decreased at the sites of first and second-order pericytes that expressed CYP4A. Capillary diameters were unaffected at the sites of other pericytes that did not express CYP4A. These findings implicate 20-HETE as a paracrine mediator in various components of the neurovascular unit and are consistent with 20-HETE's emerging role in the regulation of cerebral blood flow, blood-brain barrier integrity, the pathogenesis of stroke and the vascular contributions to cognitive impairment and dementia. Moreover, this study highlights GPR75 as a potential therapeutic target for the treatment of these devastating conditions.

9.
Front Behav Neurosci ; 14: 87, 2020.
Article in English | MEDLINE | ID: mdl-32670029

ABSTRACT

Individual variance in vulnerability to develop addictions is influenced by social factors. Specifically, drug-taking in a sexual context appears to enhance further drug-seeking behavior in human users, as these users identify the effects of drugs to enhance sexual pleasure as a primary reason for continued drug use. Methamphetamine (Meth) is commonly used in this context. Similarly, male rats that self-administered Meth immediately followed by sexual behavior display enhanced drug-seeking behavior, including attenuation of extinction and increased reinstatement to seeking of Meth-associated cues. Hence, drug-taking in a sexual context enhances vulnerability for addiction. However, the neural mechanisms by which this occurs are unknown. Here the hypothesis was tested that medial prefrontal cortex is essential for this effect of Meth and sex when experienced concurrently. First it was shown that CaMKII neurons in the anterior cingulate area (ACA) were co-activated by both Meth and sex. Next, chemogenetic inactivation of ACA CaMKII cells using AAV5-CaMKIIa-hM4Di-mCherry was shown not to affect Meth-induced locomotor activity or sexual behavior. Subsequently, chemogenetic inactivation of ACA CaMKII neurons during Meth self-administration followed by sexual behavior was shown to prevent the effects of Meth and sex on enhanced reinstatement of Meth-seeking but did not affect enhanced drug-seeking during extinction tests. These results indicate that ACA CaMKII cell activation during exposure to Meth in a sexual context plays an essential role in the subsequent enhancement of drug-seeking during reinstatement tests.

10.
Endocrinology ; 161(3)2020 03 01.
Article in English | MEDLINE | ID: mdl-32067028

ABSTRACT

Elevated and sustained estradiol concentrations cause a gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH) surge that is necessary for ovulation. In sheep, several different neural systems have been implicated in this stimulatory action of estradiol and this study focused on somatostatin (SST) neurons in the ventral lateral region of the ventral medial nucleus (vlVMN) which express c-Fos during the surge. First, we determined if increased activity of SST neurons could be related to elevated GnRH secretion by assessing SST synapses onto GnRH neurons and neurons coexpressing kisspeptin, neurokinin B, dynorphin (KNDy). We found that the percentage of preoptic area GnRH neurons that receive SST input increased during the surge compared with other phases of the cycle. However, since SST is generally inhibitory, and pharmacological manipulation of SST signaling did not alter the LH surge in sheep, we hypothesized that nitric oxide (NO) was also produced by these neurons to account for their activation during the surge. In support of this hypothesis we found that (1) the majority of SST cells in the vlVMN (>80%) contained neuronal nitric oxide synthase (nNOS); (2) the expression of c-Fos in dual-labeled SST-nNOS cells, but not in single-labeled cells, increased during the surge compared with other phases of the cycle; and (3) intracerebroventricular (ICV) infusion of the nitric oxide synthase inhibitor, N(G)-nitro-L-arginine methyl ester, completely blocked the estrogen-induced LH surge. These data support the hypothesis that the population of SST-nNOS cells in the vlVMN are a source of NO that is critical for the LH surge, and we propose that they are an important site of estradiol positive feedback in sheep.


Subject(s)
Luteinizing Hormone/blood , Nitric Oxide/metabolism , Ovulation , Sheep/blood , Ventromedial Hypothalamic Nucleus/enzymology , Animals , Female , Nitric Oxide Synthase Type I/metabolism , Somatostatin/metabolism
11.
Endocrinology ; 161(4)2020 04 01.
Article in English | MEDLINE | ID: mdl-32005991

ABSTRACT

Evidence suggests that the hypothalamic-pituitary-gonadal (HPG) axis is active during the critical period for sexual differentiation of the ovine sexually dimorphic nucleus, which occurs between gestational day (GD) 60 and 90. Two possible neuropeptides that could activate the fetal HPG axis are kisspeptin and neurokinin B (NKB). We used GD85 fetal lambs to determine whether intravenous administration of kisspeptin-10 (KP-10) or senktide (NKB agonist) could elicit luteinizing hormone (LH) release. Immunohistochemistry and fluorescent in situ hybridization (FISH) were employed to localize these peptides in brains of GD60 and GD85 lamb fetuses. In anesthetized fetuses, KP-10 elicited robust release of LH that was accompanied by a delayed rise in serum testosterone in males. Pretreatment with the GnRH receptor antagonist (acyline) abolished the LH response to KP-10, confirming a hypothalamic site of action. In unanesthetized fetuses, senktide, as well as KP-10, elicited LH release. The senktide response of females was greater than that of males, indicating a difference in NKB sensitivity between sexes. Gonadotropin-releasing hormone also induced a greater LH discharge in females than in males, indicating that testosterone negative feedback is mediated through pituitary gonadotrophs. Kisspeptin and NKB immunoreactive cells in the arcuate nucleus were more abundant in females than in males. Greater than 85% of arcuate kisspeptin cells costained for NKB. FISH revealed that the majority of these were kisspeptin/NKB/dynorphin (KNDy) neurons. These results support the hypothesis that kisspeptin-GnRH signaling regulates the reproductive axis of the ovine fetus during the prenatal critical period acting to maintain a stable androgen milieu necessary for brain masculinization.


Subject(s)
Hypothalamus/drug effects , Kisspeptins/pharmacology , Luteinizing Hormone/blood , Testosterone/blood , Animals , Female , Fetus , Gonadotropin-Releasing Hormone/pharmacology , Hypothalamus/metabolism , Kisspeptins/metabolism , Male , Neurokinin B/metabolism , Oligopeptides/pharmacology , Peptide Fragments/pharmacology , Pregnancy , Receptors, Kisspeptin-1/agonists , Receptors, Neurokinin-3/agonists , Sheep , Substance P/analogs & derivatives , Substance P/pharmacology
12.
Semin Reprod Med ; 37(2): 71-83, 2019 03.
Article in English | MEDLINE | ID: mdl-31847027

ABSTRACT

There is now considerable evidence supporting the role of a subpopulation of neurons in the arcuate nucleus of the hypothalamus that coexpress kisspeptin, neurokinin B, and dynorphin (abbreviated as KNDy neurons) as the long sought-after gonadotropin-releasing hormone (GnRH) pulse generator. The "KNDy hypothesis" of pulse generation has largely been based on findings in rodents and ruminants, and there is considerably less information about the anatomical and functional organization of the KNDy subpopulation in the primate hypothalamus. In this review, we focus on the applicability of this hypothesis, and the roles of kisspeptin, neurokinin B, and dynorphin in reproduction, to humans and nonhuman primates, reviewing available data and pointing out important gaps in our current knowledge. With recent application of drugs that target KNDy peptides and their receptors to therapeutic treatments for reproductive disorders, it is imperative we fully understand the primate KNDy network and its role in the control of GnRH secretion, as well as species differences in this system that may exist between humans, nonhuman primates, and other mammals.


Subject(s)
Dynorphins/metabolism , Gonadotropin-Releasing Hormone/metabolism , Kisspeptins/metabolism , Neurokinin B/metabolism , Animals , Female , Haplorhini , Humans , Hypothalamus , Male
13.
Sci Rep ; 9(1): 14768, 2019 10 14.
Article in English | MEDLINE | ID: mdl-31611573

ABSTRACT

Neurons in the hypothalamic arcuate nucleus (ARC) that co-express kisspeptin, neurokinin B and dynorphin (KNDy cells) are essential for mammalian reproduction as key regulators of gonadotropin-releasing hormone (GnRH) secretion. Although multiple endogenous and exogenous signals act indirectly via KNDy neurons to regulate GnRH, the identity of upstream neurons that provide synaptic input to this subpopulation is unclear. We used rabies-mediated tract-tracing in transgenic Kiss1-Cre mice combined with whole-brain optical clearing and multiple-label immunofluorescence to create a comprehensive and quantitative brain-wide map of neurons providing monosynaptic input to KNDy cells, as well as identify the estrogen receptor content and peptidergic phenotype of afferents. Over 90% of monosynaptic input to KNDy neurons originated from hypothalamic nuclei in both male and female mice. The greatest input arose from non-KNDy ARC neurons, including proopiomelanocortin-expressing cells. Significant female-dominant sex differences in afferent input were detected from estrogen-sensitive hypothalamic nuclei critical for reproductive endocrine function and sexual behavior in mice, indicating KNDy cells may provide a unique site for the coordination of sex-specific behavior and gonadotropin release. These data provide key insight into the structural framework underlying the ability of KNDy neurons to integrate endogenous and environmental signals important for the regulation of reproductive function.


Subject(s)
Dynorphins/metabolism , Hypothalamus/physiology , Kisspeptins/metabolism , Neurokinin B/metabolism , Animals , Female , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/cytology , Male , Mice , Neurons/cytology , Neurons/metabolism , Reproduction , Sex Characteristics
14.
Endocrinology ; 160(12): 2990-3000, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31599937

ABSTRACT

Recent evidence has implicated neurokinin B (NKB) signaling in the retrochiasmatic area (RCh) of the ewe in the LH surge. To test this hypothesis, we first lesioned NK3R neurons in this area by using a saporin conjugate (NK3-SAP). Three weeks after bilateral injection of NK3-SAP or a blank control (BLK-SAP) into the RCh, an LH surge was induced by using an artificial follicular-phase model in ovariectomized ewes. NK3-SAP lesioned approximately 88% of RCh NK3R-containing neurons and reduced the amplitude of the estrogen-induced LH surge by 58%, an inhibition similar to that seen previously with intracerebroventricular (icv) infusion of a KISS1R antagonist (p271). We next tested the hypothesis that NKB signaling in the RCh acts via kisspeptin by determining whether the combined effects of NK3R-SAP lesions and icv infusion of p271 were additive. Experiment 1 was replicated except that ewes received two sequential artificial follicular phases with infusions of p271 or vehicle using a crossover design. The combination of the two treatments decreased the peak of the LH surge by 59%, which was similar to that seen with NK3-SAP (52%) or p271 (54%) alone. In contrast, p271 infusion delayed the onset and peak of the LH surge in both NK3-SAP- and BLK-SAP-injected ewes. Based on these data, we propose that NKB signaling in the RCh increases kisspeptin levels critical for the full amplitude of the LH surge in the ewe but that kisspeptin release occurs independently of RCh input at the onset of the surge to initiate GnRH secretion.


Subject(s)
Hypothalamus/metabolism , Kisspeptins/metabolism , Luteinizing Hormone/metabolism , Neurokinin B/metabolism , Animals , Female , Sheep
15.
Endocrinology ; 160(11): 2529-2542, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31415088

ABSTRACT

Prenatal testosterone (T)-treated female sheep display reproductive deficits similar to women with polycystic ovarian syndrome (PCOS), including an increase in LH pulse frequency due to actions of the central GnRH pulse generator. In this study, we used multiple-label immunocytochemistry to investigate the possibility of changes in the γ-aminobutyric acid (GABA) neurotransmitter system at two key components of the GnRH pulse generator in prenatal T-treated sheep: kisspeptin/neurokinin B/dynorphin (KNDy) neurons of the arcuate nucleus, and GnRH neurons in the preoptic area (POA) and mediobasal hypothalamus (MBH). We observed a significant decrease and increase, respectively, in the number of GABAergic synapses onto POA and MBH GnRH neurons in prenatal T-treated ewes; additionally, there was a significant increase in the number of GABAergic inputs onto KNDy neurons. To determine the actions of GABA on GnRH and KNDy neurons, we examined colocalization with the chloride transporters NKCC1 and KCC2, which indicate stimulatory or inhibitory activation of neurons by GABA, respectively. Most GnRH neurons in both POA and MBH colocalized NKCC1 cotransporter whereas none contained the KCC2 cotransporter. Most KNDy neurons colocalized either NKCC1 or KCC2, and 28% of the KNDy population contained NKCC1 alone. Therefore, we suggest that, as in the mouse, GABA in the sheep is stimulatory to GnRH neurons, as well as to a subset of KNDy neurons. Increased numbers of stimulatory GABAergic inputs to both MBH GnRH and KNDy neurons in prenatal T-treated animals may contribute to alterations in steroid feedback control and increased GnRH/LH pulse frequency seen in this animal model of PCOS.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiopathology , GABAergic Neurons/physiology , Gonadotropin-Releasing Hormone/metabolism , Polycystic Ovary Syndrome/physiopathology , Preoptic Area/physiopathology , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Disease Models, Animal , Dynorphins/metabolism , Female , Kisspeptins/metabolism , Neurokinin B/metabolism , Polycystic Ovary Syndrome/metabolism , Pregnancy , Prenatal Exposure Delayed Effects , Preoptic Area/metabolism , Sheep , Solute Carrier Family 12, Member 2/metabolism , Symporters/metabolism , Testosterone , K Cl- Cotransporters
16.
J Neurotrauma ; 36(24): 3378-3393, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31111794

ABSTRACT

Spinal cord injury (SCI) causes sexual dysfunction, including anejaculation in men. Likewise, chronic mid-thoracic contusion injury impairs ejaculatory reflexes in male rats. Ejaculation is controlled by a spinal ejaculation generator (SEG) comprised of a population of lumbar spinothalamic (LSt) neurons. LSt neurons co-express four neuropeptides, including gastrin-releasing peptide (GRP) and galanin and control ejaculation via release of these peptides in lumbar and sacral autonomic and motor nuclei. Here, we tested the hypothesis that contusion injury causes a disruption of the neuropeptides that are expressed in LSt cell bodies and axon terminals, thereby causing ejaculatory dysfunction. Male Sprague Dawley rats received contusion or sham surgery at spinal levels T6-7. Five to six weeks later, animals were perfused and spinal cords were immunoprocessed for galanin and GRP. Results showed that numbers of cells immunoreactive for galanin were not altered by SCI, suggesting that LSt cells are not ablated by SCI. In contrast, GRP immunoreactivity was decreased in LSt cells following SCI, evidenced by fewer GRP and galanin/GRP dual labeled cells. However, SCI did not affect efferent connections of LSt, cells as axon terminals containing galanin or GRP in contact with autonomic cells were not reduced following SCI. Finally, no changes in testosterone plasma levels or androgen receptor expression were noted after SCI. In conclusion, chronic contusion injury decreased immunoreactivity for GRP in LSt cell soma, but did not affect LSt neurons per se or LSt connections within the SEG. Since GRP is essential for triggering ejaculation, such loss may contribute to ejaculatory dysfunction following SCI.


Subject(s)
Ejaculation/physiology , Gastrin-Releasing Peptide/metabolism , Sexual Dysfunction, Physiological/metabolism , Spinal Cord Injuries/metabolism , Animals , Chronic Disease , Gastrin-Releasing Peptide/analysis , Locomotion/physiology , Male , Rats , Rats, Sprague-Dawley , Sexual Dysfunction, Physiological/physiopathology , Spinal Cord Injuries/physiopathology , Thoracic Vertebrae/injuries
17.
Neuropsychopharmacology ; 44(3): 503-513, 2019 02.
Article in English | MEDLINE | ID: mdl-30337639

ABSTRACT

Vulnerability to develop addiction is influenced by numerous factors, including social behavior. Specifically, in human users, drug taking in a socio-sexual context appears to enhance further drug-seeking behavior. Users report heightened sexual pleasure as a motivation for further drug use and display risk behaviors even when tested in drug-free state. Here, using a preclinical model of limited voluntary drug use in rats, the hypothesis was tested that methamphetamine (Meth)-taking concurrently with socio-sexual experience increases vulnerability to addiction. Male Sprague Dawley rats were socially housed and underwent limited-access Meth self-administration (maximum 1 mg/kg/session). Meth-taking was either concurrent or non-concurrent with sexual behavior: concurrent animals were mated with a receptive female immediately after each session, while non-concurrent animals gained equivalent sexual experience the week prior. Next, drug-seeking behaviors were measured during cue reactivity, extinction, and reinstatement sessions using different extinction and reinstatement protocols in 4 separate studies. Both groups equally acquired Meth self-administration and did not differ in total Meth intake. However, drug-seeking behavior was significantly higher in concurrent animals during cue reactivity tasks, extinction sessions, and cue- or Meth-induced reinstatement tests. In addition, sexual behavior in the absence of Meth triggered reinstatement of drug-seeking in concurrent animals. These results indicate that Meth-taking in a socio-sexual context significantly enhances vulnerability for drug addiction in male rats. This preclinical paradigm of drug self-administration concurrent with socio-sexual behavior provides a useful model for studying the underlying neurobiology of socially driven vulnerability to drug addiction.


Subject(s)
Behavior, Addictive , Central Nervous System Stimulants/administration & dosage , Methamphetamine/administration & dosage , Sexual Behavior, Animal , Social Behavior , Animals , Disease Models, Animal , Male , Rats , Rats, Sprague-Dawley , Self Administration
18.
Int Rev Neurobiol ; 140: 249-270, 2018.
Article in English | MEDLINE | ID: mdl-30193706

ABSTRACT

Many factors, including social elements, influence drug addiction in humans and can be modeled in laboratory rodents. In general, the presence of social reward is protective against drug abuse and the absence or removal of social reward in both humans and rodents increases vulnerability to drug addiction. The current review chapter is focused on studies from our lab that have examined the effects of sociosexual behavior in male rats on drug-induced behaviors, including changes in both psychostimulant and opiate behavior. Furthermore, we review the underlying neural mechanisms by which these effects occur. Together, these results may help elucidate the neural mechanisms underlying the interaction between social and drug rewards and the mechanisms by which a loss of social rewards increase the vulnerability to drug addiction development.


Subject(s)
Behavior, Addictive/physiopathology , Behavior, Animal/physiology , Central Nervous System Stimulants/pharmacology , Dopaminergic Neurons/metabolism , Nucleus Accumbens/metabolism , Opiate Alkaloids/pharmacology , Reinforcement, Psychology , Sexual Behavior, Animal/physiology , Signal Transduction/physiology , Social Behavior , Ventral Tegmental Area/metabolism , Animals , Behavior, Animal/drug effects , Dopaminergic Neurons/cytology , Dopaminergic Neurons/drug effects , Male , Nucleus Accumbens/cytology , Nucleus Accumbens/drug effects , Rats , Signal Transduction/drug effects , Ventral Tegmental Area/cytology , Ventral Tegmental Area/drug effects
19.
Endocrinology ; 159(9): 3219-3234, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30010844

ABSTRACT

In the past decade since kisspeptin/neurokinin B/dynorphin (KNDy) cells were first identified in the mammalian hypothalamus, a plethora of new research has emerged adding insights into the role of this neuronal population in reproductive neuroendocrine function, including the basis for GnRH pulse generation and the mechanisms underlying the steroid feedback control of GnRH secretion. In this mini-review, we provide an update of evidence regarding the roles of KNDy peptides and their postsynaptic receptors in producing episodic GnRH release and assess the relative contribution of KNDy neurons to the "GnRH pulse generator." In addition, we examine recent work investigating the role of KNDy neurons as mediators of steroid hormone negative feedback and review evidence for their involvement in the preovulatory GnRH/LH surge, taking into account species differences that exist among rodents, ruminants, and primates. Finally, we summarize emerging roles of KNDy neurons in other aspects of reproductive function and in nonreproductive functions and discuss critical unresolved questions in our understanding of KNDy neurobiology.


Subject(s)
Dynorphins/metabolism , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Kisspeptins/metabolism , Neurokinin B/metabolism , Neurons/metabolism , Animals , Estrogens/metabolism , Female , Humans , Hypothalamus/cytology , Hypothalamus/physiology , Neurons/physiology , Progesterone/metabolism , Reproduction/physiology , Testosterone/metabolism
20.
Endocrinology ; 159(9): 3187-3199, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30016419

ABSTRACT

A subpopulation of neurons located within the arcuate nucleus, colocalizing kisspeptin, neurokinin B, and dynorphin (Dyn; termed KNDy neurons), represents key mediators of pulsatile GnRH secretion. The KNDy model of GnRH pulse generation proposes that Dyn terminates each pulse. However, it is unknown where and when during a pulse that Dyn is released to inhibit GnRH secretion. Dyn acts via the κ opioid receptor (KOR), and KOR is present in KNDy and GnRH neurons in sheep. KOR, similar to other G protein-coupled receptors, are internalized after exposure to ligand, and thus internalization can be used as a marker of endogenous Dyn release. Thus, we hypothesized that KOR will be internalized at pulse termination in both KNDy and GnRH neurons. To test this hypothesis, GnRH pulses were induced in gonad-intact anestrous ewes by injection of neurokinin B (NKB) into the third ventricle and animals were euthanized at times of either pulse onset or termination. NKB injections produced increased internalization of KOR within KNDy neurons during both pulse onset and termination. In contrast, KOR internalization into GnRH neurons was seen only during pulse termination, and only in GnRH neurons within the mediobasal hypothalamus (MBH). Overall, our results indicate that Dyn is released onto KNDy cells at the time of pulse onset, and continues to be released during the duration of the pulse. In contrast, Dyn is released onto MBH GnRH neurons only at pulse termination and thus actions of Dyn upon KNDy and GnRH cell bodies may be critical for pulse termination.


Subject(s)
Dynorphins/metabolism , Gonadotropin-Releasing Hormone/metabolism , Neurons/metabolism , Receptors, Opioid, kappa/metabolism , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/metabolism , Dynorphins/drug effects , Female , Hypothalamus/cytology , Hypothalamus/metabolism , Kisspeptins/metabolism , Neurokinin B/metabolism , Neurokinin B/pharmacology , Neurons/drug effects , Receptors, Opioid, kappa/drug effects , Sheep
SELECTION OF CITATIONS
SEARCH DETAIL
...