Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
2.
Domest Anim Endocrinol ; 68: 83-91, 2019 07.
Article in English | MEDLINE | ID: mdl-30908995

ABSTRACT

Pulsatile gonadotropin-releasing hormone (GnRH) secretion is indispensable for reproduction in mammals. Kisspeptin neurons in the hypothalamic arcuate nucleus (ARC), referred to as KNDy neurons because of the coexpression of neurokinin B and dynorphin A, are considered as components of the GnRH pulse generator that produces rhythmic GnRH secretion. The present study aimed to investigate if peripheral administration of PF-4455242, a κ-opioid receptor (KOR, a dynorphin A receptor) antagonist, facilitates pulsatile luteinizing hormone (LH) secretion and GnRH pulse generator activity in estrogen-treated ovariectomized Shiba goats to determine the possibility of using KOR antagonists to artificially control ovarian activities. PF-4455242 was intravenously infused for 4 h (1 or 10 µmol/kg body weight/4 h) or as a single subcutaneous injection (1 or 10 µmol/kg body weight). In a separate experiment, the same KOR antagonist (10 µmol/kg body weight/4 h) was intravenously infused during the recording of multiple unit activity (MUA) in the ARC that reflects the activity of the GnRH pulse generator to test the effects of KOR antagonist administration on GnRH pulse generator activity. Intravenous infusion and single subcutaneous injection of the KOR antagonist significantly increased the frequency of LH pulses compared with controls. Intravenous infusion of KOR antagonist also significantly increased the frequency of episodic bursts in the MUA. The present study demonstrates that peripherally administered KOR antagonist stimulates pulsatile LH secretion by acting on the GnRH pulse generator, and peripheral administration of PF-4455242 can be used to facilitate pulsatile LH secretion, which in turn facilitates ovarian activities in farm animals.


Subject(s)
Biphenyl Compounds/pharmacology , Estrogens/administration & dosage , Goats/physiology , Gonadotropin-Releasing Hormone/metabolism , Receptors, Opioid, kappa/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Biphenyl Compounds/administration & dosage , Female , Gene Expression Regulation/drug effects , Injections, Intravenous , Injections, Subcutaneous , Ovariectomy/veterinary , Sulfonamides/administration & dosage
3.
J Neuroendocrinol ; 29(8)2017 08.
Article in English | MEDLINE | ID: mdl-28699305

ABSTRACT

Olfactory stimuli play an important role in regulating reproductive functions in mammals. The present study investigated the effect of olfactory signals derived from male rats on kisspeptin neuronal activity and luteinising hormone (LH) secretion in female rats. Wistar-Imamichi strain female rats were ovariectomised (OVX) and implanted with preovulatory levels of 17ß-oestradiol (E2 ). OVX+E2 rats were killed 1 hour after exposure to either: clean bedding, female-soiled bedding or male-soiled bedding. Dual staining for Kiss1 mRNA in situ hybridisation and c-Fos immunohistochemistry revealed that the numbers of Kiss1-expressing cells and c-Fos-immunopositive Kiss1-expressing cells in the anteroventral periventricular nucleus (AVPV) were significantly higher in OVX+E2 rats exposed to male-soiled bedding than those of the other groups. No significant difference was found with respect to the number of c-Fos-immunopositive Kiss1-expressing cells in the arcuate nucleus and c-Fos-immunopositive Gnrh1-expressing cells between the groups. The number of c-Fos-immunopositive cells was also significantly higher in the limbic system consisting of several nuclei, such as the bed nucleus of the stria terminalis, the cortical amygdala and the medial amygdala, in OVX+E2 rats exposed to male-soiled bedding than the other groups. OVX+E2 rats exposed to male-soiled bedding showed apparent LH surges, and the peak of the LH surge and area under the curve of LH concentrations in the OVX+E2 group were significantly higher than those of the other two groups. These results suggest that olfactory signals derived from male rats activate AVPV kisspeptin neurones, likely via the limbic system, resulting in enhancement of the peak of the LH surge in female rats. Taken together, the results of the present study suggests that AVPV kisspeptin neurones are a target of olfactory signals to modulate LH release in female rats.


Subject(s)
Hypothalamus, Anterior/metabolism , Kisspeptins/metabolism , Luteinizing Hormone/metabolism , Neurons/metabolism , Pheromones/physiology , Animals , Brain/metabolism , Estradiol/administration & dosage , Female , Male , Ovariectomy , Pheromones/administration & dosage , Rats, Wistar
4.
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
5.
J Neuroendocrinol ; 28(10)2016 10.
Article in English | MEDLINE | ID: mdl-27344056

ABSTRACT

Rodents show apparent sex differences in their sexual behaviours. The present study used Kiss1 knockout (KO) rats to evaluate the role of kisspeptin in the defeminisation/masculinisation of the brain mechanism that controls sexual behaviours. Castrated adult Kiss1 KO males treated with testosterone showed no male sexual behaviours but demonstrated the oestrogen-induced lordosis behaviours found in wild-type females. The sizes of some of the sexual dimorphic nuclei of Kiss1 KO male rats are similar to those of females. Plasma testosterone levels at embryonic day 18 and postnatal day 0 (PND0) in Kiss1 KO males were high, similar to wild-type males, indicating that perinatal testosterone is secreted in a kisspeptin-independent manner. Long-term exposure to testosterone from peripubertal to adult periods restored mounts and intromissions in KO males, suggesting that kisspeptin-dependent peripubertal testosterone secretion is required to masculinise the brain mechanism. This long-term testosterone treatment failed to abolish lordosis behaviours in KO males, whereas kisspeptin replacement at PND0 reduced lordosis quotients in Kiss1 KO males but not in KO females. These results suggest that kisspeptin itself is required to defeminise behaviour in the perinatal period, in cooperation with testosterone. Oestradiol benzoate treatment at PND0 suppressed lordosis quotients in Kiss1 KO rats, indicating that the mechanisms downstream of oestradiol work properly in the absence of kisspeptin. There was no significant difference in aromatase gene expression in the whole hypothalamus between Kiss1 KO and wild-type male rats at PND0. Taken together, the present study demonstrates that both perinatal kisspeptin and kisspeptin-independent testosterone are required for defeminisation of the brain, whereas kisspeptin-dependent testosterone during peripuberty to adulthood is needed for masculinisation of the brain in male rats.


Subject(s)
Brain/physiology , Kisspeptins/physiology , Sex Differentiation , Testosterone/physiology , Animals , Animals, Newborn , Brain/drug effects , Castration , Female , Gene Knockout Techniques , Kisspeptins/genetics , Male , Sex Characteristics , Sex Differentiation/drug effects , Sexual Behavior, Animal , Testosterone/administration & dosage , Testosterone/blood
6.
J Neuroendocrinol ; 27(3): 187-97, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25582792

ABSTRACT

Kisspeptin, encoded by the Kiss1 gene, has attracted attention as a key candidate neuropeptide in controlling puberty and reproduction via regulation of gonadotrophin-releasing hormone (GnRH) secretion in mammals. Pioneer studies with Kiss1 or its cognate receptor Gpr54 knockout (KO) mice showed the indispensable role of kisspeptin-GPR54 signalling in the control of animal reproduction, although detailed analyses of gonadotrophin secretion, especially pulsatile and surge-mode of luteinising hormone (LH) secretion, were limited. Thus, in the present study, we have generated Kiss1 KO rats aiming to evaluate a key role of kisspeptin in governing reproduction via pulse and surge modes of GnRH/LH secretion. Kiss1 KO male and female rats showed a complete suppression of pulsatile LH secretion, which is responsible for folliculogenesis and spermatogenesis, and an absence of puberty and atrophic gonads. Kiss1 KO female rats showed no spontaneous LH/follicle-stimulating hormone surge and an oestrogen-induced LH surge, suggesting that the GnRH surge generation system, which is responsible for ovulation, does not function without kisspeptin. Furthermore, challenge of major stimulatory neurotransmitters, such as monosodium glutamate, NMDA and norepinephrine, failed to stimulate LH secretion in Kiss1 KO rats, albeit they stimulated LH release in wild-type controls. Taken together, the results of the present study confirm that kisspeptin plays an indispensable role in generating two modes (pulse and surge) of GnRH/gonadotrophin secretion to regulate puberty onset and normal reproductive performance. In addition, the present study suggests that kisspeptin neurones play a critical role as a hub integrating major stimulatory neural inputs to GnRH neurones, using newly established Kiss1 KO rats, which serve as a useful model for detailed analysis of hormonal profiles.


Subject(s)
Glutamic Acid/physiology , Kisspeptins/physiology , Luteinizing Hormone/metabolism , Sexual Maturation/physiology , Animals , Female , Follicle Stimulating Hormone/metabolism , Kisspeptins/genetics , Male , Mice, Knockout , N-Methylaspartate/physiology , Norepinephrine/physiology , Rats , Sexual Maturation/genetics
7.
J Neuroendocrinol ; 27(1): 57-65, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25367275

ABSTRACT

A luteinising hormone (LH) surge is fundamental to the induction of ovulation in mammalian females. The administration of a preovulatory level of oestrogen evokes an LH surge in ovariectomised females, whereas the response to oestrogen in castrated males differs among species; namely, the LH surge-generating system is sexually differentiated in some species (e.g. rodents and sheep) but not in others (e.g. primates). In the present study, we aimed to determine whether there is a functional LH surge-generating system in male goats, and whether hypothalamic kisspeptin neurones in male goats are involved in the regulation of surge-like LH secretion. By i.v. infusion of oestradiol (E2; 6 µg/h) for 16 h, a surge-like LH increase occurred in both castrated male and ovariectomised female goats, although the mean peak LH concentration was lower and the mean peak of the LH surge was later in males compared to females. Dual staining with KISS1 in situ hybridisation and c-Fos immunohistochemistry revealed that E2 treatment significantly increased c-Fos expression in the medial preoptic area (mPOA) KISS1 cells in castrated males, as well as ovariectomised females. By contrast, dual-labelled cells were scarcely detected in the arcuate nucleus (ARC) after E2 treatment in both sexes. These data suggest that kisspeptin neurones in the mPOA, but not those in the ARC, are involved in the induction of surge-like LH secretion in both male and female goats. In summary, our data show that the mechanism that initiates the LH surge in response to oestrogen, the mPOA kisspeptin neurones, is functional in male goats. Thus, sexual differentiation of the LH surge-generating system would not be applicable to goats.


Subject(s)
Kisspeptins/metabolism , Luteinizing Hormone/biosynthesis , Neurons/metabolism , Preoptic Area/metabolism , Animals , Female , Goats , In Situ Hybridization , Kisspeptins/genetics , Luteinizing Hormone/blood , Male , Preoptic Area/cytology
8.
J Neuroendocrinol ; 26(12): 909-17, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25283748

ABSTRACT

The oestrogen-induced luteinising hormone (LH) surge is evident in male primates, including humans, whereas male rodents never show the LH surge, even when treated with a preovulatory level of oestrogen. This suggests that the central mechanism governing reproductive hormones in primates is different from that in rodents. The present study aimed to investigate whether male Japanese monkeys conserve a brain mechanism mediating the oestrogen-induced LH surge via activation of kisspeptin neurones. Adult male and female Japanese monkeys were gonadectomised and then were treated with oestradiol-17ß for 2 weeks followed by a bolus injection of oestradiol benzoate. Both male and female monkeys showed an oestrogen-induced LH surge. In gonadectomised monkeys sacrificed just before the anticipated time of the LH surge, oestrogen treatment significantly increased the number of KISS1-expressing cells in the preoptic area (POA) and enhanced the expression of c-fos in POA KISS1-positive cells of males and females. The oestrogen treatment failed to induce c-fos expression in the arcuate nucleus (ARC) kisspeptin neurones in both sexes just prior to LH surge onset. Thus, kisspeptin neurones in the POA but not in the ARC might be involved in the positive-feedback action of oestrogen that induces LH surge in male Japanese monkeys, as well as female monkeys. The present results indicate that oestrogen-induced activation of POA kisspeptin neurones may contribute to the LH surge generation in both sexes. The conservation of the LH surge generating system found in adult male primates, unlike rodents, could be a result of the capability of oestrogen to induce POA kisspeptin expression and activation.


Subject(s)
Estradiol/analogs & derivatives , Kisspeptins/metabolism , Luteinizing Hormone/blood , Neurons/drug effects , Preoptic Area/cytology , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/physiology , Estradiol/blood , Estradiol/pharmacology , Female , Kisspeptins/biosynthesis , Macaca , Male , Neurons/metabolism , Neurons/physiology , Preoptic Area/drug effects , Preoptic Area/physiology
9.
J Neuroendocrinol ; 25(3): 251-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22994299

ABSTRACT

Female rats show a gonadotrophin-releasing hormone (GnRH)/luteinising hormone (LH) surge in the presence of a preovulatory level of oestrogen, whereas males do not because of brain defeminisation during the developmental period by perinatal oestrogen converted from androgen. The present study aimed to identify the site(s) of oestrogen action and the critical period for defeminising the mechanism regulating the GnRH/LH surge. Animals given perinatal treatments, such as steroidal manipulations, brain local implantation of oestradiol (E(2) ) or administration of an NMDA antagonist, were examined for their ability to show an E(2) -induced LH surge at adulthood. Lordosis behaviour was examined to compare the mechanisms defeminising the GnRH/LH surge and sexual behaviour. A single s.c. oestradiol-benzoate administration on either the day before birth (E21), the day of birth (D0) or day 5 (D5) postpartum completely abolished the E(2) -induced LH surge at adulthood in female rats, although the same treatment did not inhibit lordosis. Perinatal castration on E21 or D0 partially rescued the E2-induced LH surge in genetically male rats, whereas castration from E21 to D5 totally rescued lordosis. Neonatal E(2) implantation in the anterior hypothalamus including the anteroventral periventricular nucleus (AVPV)/preoptic area (POA) abolished the E(2) -induced LH surge in female rats, whereas E(2) implantation in the mid and posterior hypothalamic regions had no inhibitory effect on the LH surge. Lordosis was not affected by neonatal E(2) implantation in any hypothalamic regions. In male rats, neonatal NMDA antagonist treatment rescued lordosis but not the LH surge. Taken together, these results suggest that an anterior hypothalamic region such as the AVPV/POA region is a perinatal site of oestrogen action where the GnRH/LH regulating system is defeminised to abolish the oestrogen-induced surge. The mechanism for defeminisation of the GnRH/LH surge system might be different from that of sexual behaviour, in terms of the site(s) of oestrogen action and critical period, as well as the neurotransmitter system involved.


Subject(s)
Estradiol/physiology , Hypothalamus/physiopathology , Lordosis/physiopathology , Luteinizing Hormone/metabolism , Animals , Animals, Newborn , Female , Male , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Sexual Behavior, Animal
10.
J Neuroendocrinol ; 24(9): 1234-42, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22536815

ABSTRACT

Follicular development and ovulation are strongly suppressed during lactation in mammals via a profound suppression of gonadotrophin secretion. The present study aimed to examine the role of oestrogen feedback action in suppressing luteinising hormone (LH) secretion and hypothalamic kisspeptin expression during the latter half of lactation. Plasma LH concentrations kept at low levels throughout the lactating period in intact and oestrogen-replaced ovariectomised (OVX) lactating rats, whereas plasma LH concentrations gradually elevated from day 10 postpartum in lactating OVX rats. OVX lactating rats showed frequent LH pulses at late lactation, although the LH pulses were significantly inhibited by an oestrogen replacement, which is much less effective on LH release in nonlactating rats. Oestrogen replacement in lactating OVX rats significantly reduced the number of Kiss1 mRNA-expressing cells in the arcuate nucleus (ARC) at late lactation, although the same oestrogen treatment did not affect the number of Kiss1-expressing cells in nonlactating controls. Exogenous kisspeptin challenge (0.2 nmol) into the third cerebroventricle significantly increased LH secretion in lactating OVX, lactating OVX + subcutaneous 17ß-oestradiol and intact lactating rats at day 16 postpartum. These results suggest that LH pulse suppression during late lactation could be a result of the enhanced oestrogen-dependent suppression of ARC kisspeptin expression.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Estradiol/physiology , Kisspeptins/biosynthesis , Lactation/metabolism , Lactation/physiology , Luteinizing Hormone/metabolism , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Estradiol/blood , Estradiol/pharmacology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Injections, Intraventricular , Kisspeptins/administration & dosage , Kisspeptins/pharmacology , Luteinizing Hormone/blood , Midline Thalamic Nuclei/drug effects , Midline Thalamic Nuclei/metabolism , Ovariectomy/methods , Rats , Rats, Wistar , Time Factors
11.
J Neuroendocrinol ; 23(10): 863-70, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21815953

ABSTRACT

The present study was conducted to determine the morphological and functional interaction between kisspeptin and gonadotrophin-releasing hormone (GnRH) neuronal elements at the median eminence in female rats to clarify a possibility that kisspeptin directly stimulates GnRH release at the nerve end. A dual immunoelectron microscopic study of kisspeptin and GnRH showed that the kisspeptin-immunoreactive nerve element directly abutted the GnRH-immunoreactive nerve element, although no obvious synaptic structure was found between kisspeptin and GnRH neurones in the median eminence. The current retrograde tracing study with FluoroGold (FG) indicates that kisspeptin neurones are not in contact with fenestrated capillaries because no FG signal was found in kisspeptin neurones when the FG was injected peripherally. This peripheral FG injection revealed the neuroendocrine neurones projecting to the median eminence because FG-positive GnRH neuronal cell bodies were found in the preoptic area. Synthetic rat kisspeptin (1-52)-amide stimulated GnRH release from the median eminence tissues in a dose-dependent manner. Thus, the present results suggest that kisspeptin at least partly exerts stimulatory effects on GnRH release from the neuronal terminals of GnRH neurones by axo-axonal nonsynaptic interaction in the median eminence.


Subject(s)
Axons , Gonadotropin-Releasing Hormone/metabolism , Kisspeptins/metabolism , Median Eminence/metabolism , Animals , Female , Immunohistochemistry , Median Eminence/ultrastructure , Radioimmunoassay , Rats , Rats, Wistar
12.
J Neuroendocrinol ; 21(10): 813-21, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19678868

ABSTRACT

Pulsatile release of gonadotrophin-releasing hormone (GnRH) is indispensable to maintain normal gonadotrophin secretion. The pulsatile secretion of GnRH is associated with synchronised electrical activity in the mediobasal hypothalamus (i.e. multiple unit activity; MUA), which is considered to reflect the rhythmic oscillations in the activity of the neuronal network that drives pulsatile GnRH secretion. However, the cellular source of this ultradian rhythm in GnRH activity is unknown. Direct input from kisspeptin neurones in the arcuate nucleus (ARC) to GnRH cell bodies in the medial preoptic area or their terminals in the median eminence could be the intrinsic source for driving the GnRH pulse generator. To determine whether kisspeptin signalling could be responsible for producing pulsatile GnRH secretion, we studied goats, measured plasma levels of luteinising hormone (LH) and recorded MUA in the posterior ARC, where the majority of kisspeptin neuronal cell bodies are located. Rhythmic volleys of MUA were found to be accompanied by LH pulses with regular intervals in the ARC, where kisspeptin neuronal cell bodies were found. Exogenous administration of kisspeptin stimulated a sustained increase in LH secretion, without influencing MUA, suggesting that the GnRH pulse generator, as reflected by MUA, originated from outside of the network of GnRH neurones, and could plausibly reflect the pacemaker activity of kisspeptin neurones, whose projections reach the median eminence where GnRH fibres project. These observations suggest that the kisspeptin neurones in the ARC may be the intrinsic source of the GnRH pulse generator.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiology , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/physiology , Neurons/physiology , Periodicity , Amino Acid Sequence , Animals , Electrodes, Implanted , Goats , Humans , Immunohistochemistry , In Situ Hybridization , Kisspeptins , Luteinizing Hormone/blood , Male , Molecular Sequence Data , Neural Pathways/physiology , Orchiectomy , Sequence Homology, Amino Acid , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
13.
J Neuroendocrinol ; 21(6): 527-37, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19500223

ABSTRACT

Kisspeptin, a peptide encoded by the Kiss1 gene, has been considered as a potential candidate for a factor triggering the onset of puberty, and its expression in the hypothalamus was found to increase during peripubertal period in rodent models. The present study aimed to clarify the oestrogenic regulation of peripubertal changes in Kiss1 mRNA expression in the anteroventral periventricular nucleus (AVPV) and hypothalamic arcuate nucleus (ARC), and to determine which population of kisspeptin neurones shows a change in kisspeptin expression parallel to that in luteinising hormone (LH) pulses at the peripubertal period. Quantitative reverse transcriptase-polymerase chain reaction and immunohistochemistry revealed an apparent increase in the ARC Kiss1 mRNA expression and kisspeptin immunoreactivity around the time of vaginal opening in intact female rats. The AVPV Kiss1 mRNA levels also increased at day 26, but decreased at day 31, and then increased at day 36/41. In ovariectomised (OVX) rats, ARC Kiss1 mRNA expression did not show peripubertal changes and was kept at a high level throughout peripubertal periods. Apparent LH pulses were found in these prepubertal OVX rats. Oestradiol replacement suppressed ARC Kiss1 mRNA expression in OVX prepubertal rats, but not in adults. Similarly, LH pulses were suppressed by oestradiol in the prepubertal period (days 21 and 26), but regular pulses were found in adulthood. The present study suggests that a pubertal increase of Kiss1/kisspeptin expression both in the ARC and AVPV is involved in the onset of puberty. These results also suggest that both LH pulses and ARC Kiss1 expression are more negatively regulated by oestrogen in prepubertal female rats compared to adult rats.


Subject(s)
Estrogens/metabolism , Gene Expression Regulation, Developmental , Hypothalamus , Proteins/metabolism , Puberty/physiology , Animals , Female , Follicle Stimulating Hormone/blood , Humans , Hypothalamus/anatomy & histology , Hypothalamus/metabolism , Kisspeptins , Luteinizing Hormone/blood , Male , Ovariectomy , Proteins/genetics , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Kisspeptin-1
14.
J Neuroendocrinol ; 21(4): 299-304, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19210293

ABSTRACT

Kisspeptin (also known as metastin), a hypothalamic peptide, has attracted attention as a key molecule in the release of gonadotrophin-releasing hormone (GnRH) in various mammalian species, such as rodents, sheep and primates. Two populations of kisspeptin neurones in the brain may control two modes of GnRH release to time the onset of puberty and regulate oestrous cyclicity in rats and mice. One population of kisspeptin neurones, located in the anteroventral periventricular nucleus, appears to be responsible for the induction of the GnRH surge that leads to the luteinising hormone surge and ovulation. The other, located in the hypothalamic arcuate nucleus, appears to be involved in generating GnRH pulses, resulting in luteinising hormone pulses followed by follicular development and steroidogenesis in the ovary. The present review focuses on the physiological role of the two populations of kisspeptin neurones in controlling gonadal functions by generating the two modes of GnRH release in a female rat model.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Gonadotropin-Releasing Hormone/metabolism , Midline Thalamic Nuclei/metabolism , Neurons/metabolism , Proteins/metabolism , Animals , Estrogens/metabolism , Female , Kisspeptins , Luteinizing Hormone/metabolism , Models, Biological , Ovary/physiology , Ovulation/physiology , Periodicity , Rats
15.
J Neuroendocrinol ; 20(5): 626-31, 2008 May.
Article in English | MEDLINE | ID: mdl-18363811

ABSTRACT

Galanin-like peptide (GALP), a ligand for three types of galanin receptor, is reported to have a role in regulating luteinising hormone (LH) release in male rodents and primates, but its role in LH release in female rodents remains controversial. The present study was conducted to test whether GALP has a stimulatory role in regulating LH secretion in female rats. The effect of i.c.v. infusion of GALP (5 nmol) on pulsatile LH release was investigated in Wistar-Imamichi strain female rats, or lean and obese Zucker rats. In oestradiol-17beta (oestradiol)-primed ovariectomised (OVX) Wistar-Imamichi female rats, i.c.v. infusion of GALP caused a gradual increase in LH release for the first 1.5 h after the infusion followed by an increased LH pulse frequency during the next 1.5 h, resulting in a significant increase in the mean LH concentrations and baseline levels of LH pulses throughout the sampling period and in the frequency of LH pulses at the last half of the period compared to vehicle-treated controls. The stimulatory effect of GALP was oestrogen-dependent because the same GALP treatment did not affect LH release in OVX rats in the absence of oestradiol. In lean Zucker rats, LH pulses were found in oestradiol-primed OVX individuals and central GALP infusion increased mean LH concentrations in the last half of the period. By contrast, few LH pulses were found in oestradiol-primed OVX obese Zucker rats reportedly with lower hypothalamic GALP expression. Central GALP infusion caused an apparent but transient increase in LH release, resulting in the significant increase in all pulse parameters of LH pulses compared to vehicle-treated controls in the first half of the sampling period. These results suggest that hypothalamic GALP is likely involved in stimulating GnRH/LH release, and that the stimulatory effect of GALP on LH release is oestrogen-dependent in female rats.


Subject(s)
Estradiol/pharmacology , Galanin-Like Peptide/pharmacology , Luteinizing Hormone/metabolism , Animals , Female , Luteinizing Hormone/blood , Obesity/blood , Ovariectomy , Pulsatile Flow/drug effects , Rats , Rats, Wistar , Rats, Zucker , Thinness/blood
16.
Endocrinology ; 148(5): 2226-32, 2007 May.
Article in English | MEDLINE | ID: mdl-17289848

ABSTRACT

Follicular development and ovulation are suppressed during lactation in various mammalian species, mainly due to the suppression of pulsatile GnRH/LH secretion. Metastin (kisspeptin-54), a KiSS-1 gene product, is an endogenous ligand for GPR54, a G-protein-coupled receptor, and suggested to play a critical role in regulating the gonadal axis. The present study therefore aims to determine whether metastin (kisspeptin-54)-GPR54 signaling in discrete brain areas is inhibited by the suckling stimulus that causes suppression of LH secretion in lactating rats. Quantitative RT-PCR revealed that the KiSS-1 mRNA level was significantly lower in the arcuate nucleus (ARC)-median eminence region in lactating ovariectomized (OVX) and estrogen-treated OVX rats than in nonlactating controls. KiSS-1 mRNA in the anteroventral periventricular nucleus was kept at a low level in both lactating and nonlactating rats despite estrogen treatment. GPR54 mRNA levels were significantly lower in lactating than nonlactating rats in the anteroventral periventricular nucleus, but the levels in lactating mothers of the preoptic area and ARC-median eminence were comparable with nonlactating controls. Although KiSS-1 mRNA-expressing cells or metastin (kisspeptin-54) immunoreactivities were densely located in the ARC of nonlactating controls, few were found in the ARC of lactating OVX animals. Various doses of metastin (kisspeptin-54) (0.02, 0.2, and 2 nmol) injected into the third ventricle caused a significant increase in LH secretion in both lactating and nonlactating OVX rats, suggesting that lactating rats are responsive to metastin (kisspeptin-54) stimulus. Thus, the present study demonstrated that KiSS-1 mRNA/metastin (kisspeptin-54) expression is inhibited in the ARC by the suckling stimulus, suggesting that the inhibition is most probably involved in suppressing LH secretion in lactating rats.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiology , Lactation/physiology , Median Eminence/physiology , Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Animals, Suckling , Arcuate Nucleus of Hypothalamus/cytology , Female , Immunohistochemistry , In Situ Hybridization , Injections, Intraventricular , Kisspeptins , Luteinizing Hormone/blood , Luteinizing Hormone/metabolism , Median Eminence/cytology , Neurons/physiology , Ovulation Inhibition/physiology , Proteins/genetics , Proteins/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Receptors, Kisspeptin-1 , Signal Transduction/physiology , Third Ventricle
17.
Oncogene ; 25(23): 3277-85, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16407821

ABSTRACT

Cyclooxygenase-2 (COX-2) plays important roles in tumor development. Especially in the early-stage colorectal tumors, COX-2 expression is often observed in the tumor stroma. However, the mechanism regulating such stromal expression of COX-2 remains unknown. In the present study, we simulated the indirect interaction between epithelial cells and stromal cells in the process of colorectal tumor development using an in vitro co-culture model in which NIH3T3 fibroblasts were co-cultured with 'sparsely' or 'densely' populated intestinal epithelial cells, Intestine-407 as a model of premalignant or benign intestinal epithelial cells, and DLD-1 and Caco-2 as models of malignant epithelial cells. COX-2 expression in NIH3T3 fibroblasts was upregulated when co-cultured with the 'dense' epithelial cells regardless of their character. Interestingly, there was pericellular hypoxia in the vicinity of NIH3T3 fibroblasts when co-cultured with 'dense' epithelial cells, and the recovery of the partial pressure of oxygen level resulted in the reduction of enhanced COX-2 expression only in NIH3T3 fibroblasts co-cultured with 'dense' Intestine-407 cells. Furthermore, COX-2 expression was also reduced by the inhibition of transcription factor AP-1. Thus, pericellular hypoxia of the stromal cells caused by densely populated epithelial cells may be one of the potent COX-2 enhancers before completion of malignant transformation during intestinal tumor development.


Subject(s)
Cyclooxygenase 2/biosynthesis , Hypoxia/enzymology , Intestinal Mucosa/cytology , Intestinal Mucosa/enzymology , Membrane Proteins/biosynthesis , Signal Transduction/physiology , Transcription Factor AP-1/physiology , Animals , Caco-2 Cells , Cell Count , Cell Line, Tumor , Coculture Techniques , Cyclooxygenase 2/physiology , Enzyme Induction/physiology , Humans , Hypoxia/pathology , Intestinal Mucosa/pathology , Membrane Proteins/physiology , Mice , NIH 3T3 Cells , Precancerous Conditions/enzymology , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Stromal Cells/enzymology , Stromal Cells/pathology
18.
Gut ; 54(10): 1437-44, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15914572

ABSTRACT

BACKGROUND AND AIMS: Although regenerating gene (REG) Ialpha protein may be involved in the inflammation and carcinogenesis in the gastrointestinal tract, its pathophysiological role in ulcerative colitis (UC) and the resulting colitic cancer remains unclear. We investigated expression of the REG Ialpha gene and its protein in UC and colitic cancer tissues. We examined whether cytokines are responsible for REG Ialpha gene expression and whether REG Ialpha protein has a trophic and/or an antiapoptotic effect on colon cancer cells. METHODS: Expression of REG Ialpha mRNA and its gene product in UC tissues was analysed by real time reverse transcription-polymerase chain reaction and immunohistochemistry, respectively. The effects of cytokines on REG Ialpha promoter activity were examined in LoVo cells by luciferase reporter assay. The effects of REG Ialpha protein on growth and H(2)O(2) induced apoptosis were examined in LoVo cells by MTT and TUNEL assays, respectively. RESULTS: REG Ialpha protein was strongly expressed in inflamed epithelium and in dysplasias and cancerous lesions in UC tissues. The level of REG Ialpha mRNA expression in UC tissues correlated significantly with severity of inflammation and disease duration. REG Ialpha promoter activity was enhanced by stimulation with interferon gamma or interleukin 6. REG Ialpha protein promoted cell growth and conferred resistance to H(2)O(2) induced apoptosis in LoVo cells. REG Ialpha protein promoted Akt phosphorylation and enhanced Bcl-xL and Bcl-2 expression in LoVo cells. CONCLUSIONS: The REG Ialpha gene is inducible by cytokines and its gene product may function as a mitogenic and/or an antiapoptotic factor in the UC-colitic cancer sequence.


Subject(s)
Colitis, Ulcerative/genetics , Colonic Neoplasms/genetics , Lithostathine/genetics , Adenoma/genetics , Adult , Aged , Aged, 80 and over , Apoptosis/genetics , Cell Division/genetics , Cell Line, Tumor , Colitis, Ulcerative/metabolism , Colonic Neoplasms/metabolism , Cytokines/genetics , Female , Gene Expression Regulation/genetics , Humans , Immunohistochemistry/methods , Intestinal Mucosa/physiology , Lithostathine/metabolism , Male , Middle Aged , Neoplasm Proteins/analysis , Proliferating Cell Nuclear Antigen/analysis , RNA, Messenger/analysis , RNA, Neoplasm/analysis
19.
Biol Reprod ; 65(6): 1634-9, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11717122

ABSTRACT

The aim of this study was to investigate the expression of cytochrome P450 aromatase (aromatase) mRNA, its activity, and estradiol-17beta (estradiol) secretion in bovine corpus luteum (CL) during the estrous cycle. Expression of aromatase mRNA was examined in CL at the early, mid, late, and regressed luteal stages by using a reverse transcription-polymerase chain reaction. Aromatase mRNA was detected in all luteal stages examined, although aromatase expression was significantly lower during the early and regressed luteal phases compared to the mid and late luteal phases. Moreover, cultured midluteal cells clearly converted exogenous [(3)H]androstenedione into estradiol, and an aromatase inhibitor significantly inhibited this conversion. To characterize the local release of estradiol within the CL during the estrous cycle, an in vitro microdialysis system (MDS) of CL was conducted. Estradiol in MDS perfusate was confirmed by a reverse-phase high-performance liquid chromatography in combination with enzyme immunoassays. Basal release of estradiol from microdialyzed CL did not change during the estrous cycle. Additionally, when freshly prepared midluteal cells were exposed to estradiol (10(-14) to 10(-9) M), estradiol stimulated prostaglandin (PG) F(2alpha) secretion (P < 0.05), although it did not affect progesterone and oxytocin secretion. The overall results indicate that estradiol is produced locally in bovine CL throughout the estrous cycle, and they suggest that estradiol plays a role in regulating PGF(2alpha) production in CL as an autocrine/paracrine factor.


Subject(s)
Aromatase/metabolism , Cattle/metabolism , Corpus Luteum/enzymology , Estradiol/biosynthesis , Androstenedione/metabolism , Animals , Aromatase/genetics , Aromatase Inhibitors , Chromatography, High Pressure Liquid , Corpus Luteum/drug effects , Corpus Luteum/metabolism , Dinoprost/metabolism , Enzyme Inhibitors/pharmacology , Estradiol/analysis , Estradiol/metabolism , Estradiol/pharmacology , Estrous Cycle , Female , Gene Expression , Luteal Phase , Microdialysis , Oxytocin/metabolism , Progesterone/metabolism , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Tritium
20.
J Vet Med Sci ; 63(12): 1303-7, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11789608

ABSTRACT

Age-related changes in immunoreactive inhibin (ir-inhibin) levels and the relationship among ir-inhibin, gonadotropins and testosterone were examined in 53 Holstein bull calves from neonates to 8.6 months old. Serum levels of ir-inhibin, luteinizing hormone (LH), follicle stimulating hormone (FSH) and testosterone, as well as ir-inhibin levels in testicular extracts, and testicular sizes were measured. All hormones were measured by specific radioimmunoassays. The concentrations of ir-inhibin in serum and testicular tissue were high in neonatal calves and tended to decrease with age. In contrast, serum concentrations of gonadotropins did not show any age-related changes within the experimental period. Serum testosterone levels and testicular sizes (length, width and weight) were positively correlated with age. Furthermore, a positive immunostaining to antiserum for the inhibin alpha-subunit was immunocytochemically observed only in Sertoli cells of the seminiferous tubules from neonates to calves less than 6 months old. These results indicate that the immature bovine testis produces and secretes high levels of ir-inhibin and that the Sertoli cells are a major source of ir-inhibin in prepubertal bull calves.


Subject(s)
Cattle/metabolism , Follicle Stimulating Hormone/blood , Inhibins/blood , Luteinizing Hormone/blood , Testis/metabolism , Testosterone/blood , Age Factors , Animals , Animals, Newborn , Cattle/blood , Cattle/growth & development , Immunohistochemistry , Male , Sertoli Cells/metabolism , Testis/cytology , Testis/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...