Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
J Oral Rehabil ; 38(3): 185-95, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20819135

ABSTRACT

The purpose of this study was to compare the speech function of low height bulb obturators with that of high height bulb obturators. Thirteen maxillectomy patients, who underwent post-operative prosthodontic rehabilitations, were included. Two obturators of the same design except for different bulb heights were fabricated for each maxillectomy patient. One of the two obturators had high bulb design and the other had low bulb design. After one of the obturators was used for a period of 3 weeks, the patient's speaking functions were evaluated by measuring nasalance scores, formant frequencies, and vowel working space areas. The same procedures were repeated with the second obturator following another 3-week period of usage. In addition, the effect of delivery sequence and anatomic conditions related to maxillectomy were analysed. The results demonstrated that the nasalance scores with the low bulb obturators were significantly higher than those with the high bulb obturators. There were no significant differences in formant frequencies based on the bulb height of the obturators. The vowel working spaces for the two obturators were similar in shape and there were no significant differences between the vowel working space areas created by the two obturators. The delivery sequence affected the results. However, there were no significant differences related to the other anatomical variables. Although low bulb obturators might function similarly with high bulb obturators in terms of the articulation of speech, they would exhibit a difficulty in controlling hypernasality in maxillectomy patients.


Subject(s)
Dental Prosthesis Design , Maxilla/surgery , Palatal Obturators , Speech/physiology , Adult , Aged , Female , Follow-Up Studies , Humans , Jaw Relation Record , Male , Maxillary Neoplasms/surgery , Middle Aged , Phonetics , Skin Transplantation , Speech Acoustics , Speech Intelligibility/physiology , Surgical Flaps , Time Factors , Voice Quality/physiology
2.
Mol Cell Biol ; 14(4): 2525-33, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8139555

ABSTRACT

We have characterized cis-acting elements involved in light regulation of the nuclear gene (GapA) encoding the A subunit of chloroplast glyceraldehyde-3-phosphate dehydrogenase (GAPDH) in Arabidopsis thaliana. Our results show that a 1.1-kb promoter fragment of the GapA gene is sufficient to confer light inducibility and organ specificity in transgenic Nicotiana tabacum (tobacco) plants, using the beta-glucuronidase gene of Escherichia coli as the reporter gene. Deletion analysis indicates that the -359 to -110 bp region of the GapA gene is necessary for light responsiveness. Within this region there are three copies of a decamer repeat (termed the Gap box) having the consensus sequence 5'-CAAATGAA(A/G)A-3', which has not been characterized in the promoter regions of other light-regulated genes. A deletion (to -247) producing loss of one copy of these elements from the GapA promoter reduces light induction by two- to threefold compared with a promoter deletion (to -359) with all three Gap boxes present, while deletion of all three Gap boxes (to -110) abolishes light induction completely. Gel mobility shift experiments using tobacco nuclei as the source of nuclear proteins show that GapA promoter fragments that contain these repeats bind strongly to a factor in the nuclear extract and that binding can be abolished by synthetic competitors consisting only of a monomer or dimer of the Gap box. Furthermore, a trimer, dimer, and monomer of the Gap box show binding activity and, like the authentic GapA promoter-derived probes, show binding activities that are correlated with Gap box copy number. These results strongly suggest that these repeats play important roles in light regulation of the GapA gene of A. thaliana.


Subject(s)
Arabidopsis/enzymology , Arabidopsis/genetics , Chloroplasts/enzymology , Gene Expression Regulation, Enzymologic/radiation effects , Genes, Plant/radiation effects , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Isoenzymes/genetics , Arabidopsis/radiation effects , Base Sequence , Cell Nucleus/metabolism , Consensus Sequence , Escherichia coli/enzymology , Escherichia coli/genetics , Genes, Bacterial , Glucuronidase/biosynthesis , Glucuronidase/genetics , Glyceraldehyde-3-Phosphate Dehydrogenases/biosynthesis , Isoenzymes/biosynthesis , Light , Macromolecular Substances , Molecular Sequence Data , Plants, Genetically Modified , Plants, Toxic , Promoter Regions, Genetic , Sequence Deletion , Nicotiana
3.
J Neuroendocrinol ; 18(10): 757-66, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16965294

ABSTRACT

There are two forms of gonadotrophin-releasing hormone (GnRH), GnRH-I and GnRH-II, in the vertebrate brain. Both GnRH-I and GnRH-II are thought to interact with the type-I GnRH receptor (GnRHR). The present study attempted to demonstrate whether GnRH-I and GnRH-II induce differential desensitisation of GnRHR and to identify the motif involved. Time course inositol phosphate (IP) accumulation assay reveals that, in cells expressing the wild-type rat GnRHR, GnRH-I induced continuous increase in IP production, whereas GnRH-II-induced IP production rate at later time points (30-120 min after ligand treatment) became attenuated. However, in cells expressing the mutant receptor in which the Ser-Glu-Pro (SEP) motif in extracellular loop 3 was replaced by Pro-Glu-Val (PEV), IP accumulation rates at later time points were more decreased by GnRH-I than GnRH-II. Ca2+ responses to repetitive GnRH applications reveal that GnRH-II desensitised the wild-type receptor faster than GnRH-I, whereas the opposite situation was observed in the PEV mutant. In addition, cell surface loss of GFP-tagged wild-type receptor was more facilitated by GnRH-II than GnRH-I, whereas that of the GFP-tagged PEV mutant receptor was more enhanced by GnRH-I than GnRH-II. The present study indicates that the SEP motif is potentially responsible for ligand species-dependent receptor desensitisation. Together, these results suggest that GnRH-I and GnRH-II may have different effects on mammalian type-I GnRHR via modulation of desensitisation rates.


Subject(s)
Amino Acid Motifs/physiology , Receptors, LHRH/physiology , Animals , Calcium/metabolism , Data Interpretation, Statistical , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Inositol Phosphates/metabolism , Ligands , Microscopy, Confocal , Mutation , Rats , Receptors, Cell Surface/metabolism , Receptors, LHRH/genetics , Species Specificity
4.
J Mol Endocrinol ; 34(3): 793-807, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15956348

ABSTRACT

Neurotensin (NT) is a tridecapeptide that functions as a neurotransmitter and neuromodulator in the nervous system. To date, three different types of NT receptor (NTR), NTR1, NTR2 and NTR3, have been identified only in mammalian species. In the present study we isolated the cDNAs for an NTR1 and a novel NTR in the bullfrog brain, designated bfNTR1 and bfNTR4 respectively. bfNTR1 and bfNTR4 encode 422- and 399-amino acid residue proteins respectively. bfNTR1 has a 64% amino acid identity with mammalian NTR1, and 34-37% identity with mammalian NTR2. bfNTR4 exhibits 43% and 45-47% identity with mammalian NTR1 and NTR2 respectively. Both receptors are mainly expressed in the brain and pituitary. bfNTR1 triggers both CRE-luc, a protein kinase A (PKA)-specific reporter, and c-fos-luc, a PKC-specific reporter, activities, indicating that bfNTR1 can activate PKA- and PKC-linked signaling pathways. However, bfNTR4 appears to be preferentially coupled to the PKA-linked pathway as it induces a higher CRE-luc activity than c-fos-luc activity. bfNTRs exhibit different pharmacological properties as compared with mammalian NTRs. Mammalian NTR1 but not NTR2 responds to NT, whereas both bfNTR1 and bfNTR4 show a high sensitivity to NT. SR 48692 and SR 142948A, antagonists for mammalian NTR1 but agonists for mammalian NTR2, function as antagonists for both bfNTR1 and bfNTR4. In conclusion, this report provides the first molecular, pharmacological and functional characterization of two NTRs in a non-mammalian vertebrate. These data should help to elucidate the phylogenetic history of the G protein-coupled NTRs in the vertebrate lineage as well as the structural features that determine their pharmacological properties.


Subject(s)
Brain/metabolism , Receptors, Neurotensin/genetics , Receptors, Neurotensin/physiology , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cloning, Molecular , DNA Primers , Ligands , Molecular Sequence Data , RNA, Messenger/genetics , Rana catesbeiana , Receptors, Neurotensin/metabolism , Sequence Homology, Amino Acid , Signal Transduction
5.
Endocrinology ; 141(2): 702-9, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10650952

ABSTRACT

Expression of pituitary adenylate cyclase-activating polypeptide (PACAP), a neuropeptide with considerable homology to vasoactive intestinal peptide, has been shown to be stimulated by gonadotropins in the ovary. The present studies further evaluated the cell-type specific expression and gonadotropin regulation of PACAP type I receptor (PACAPR) messenger RNA in immature rat ovaries and in cultured preovulatory follicles. Northern blot analysis of ovaries obtained from prepubertal rats revealed the increased expression of PACAPR during prepubertal development. The major cell types expressing PACAPR messenger RNA were granulosa cells of large preantral follicles. Treatment of immature rats with PMSG caused a decrease in ovarian PACAPR expression. In contrast, treatment with human (h) CG at 2 days after PMSG treatment stimulated ovarian PACAPR messenger RNA within 3-6 h in granulosa cells of preovulatory follicles. Treatment of cultured preovulatory follicles in vitro with LH further confirmed the time- and dose-dependent stimulation of PACAPR by gonadotropins in granulosa cells of preovulatory follicles. Moreover, RNase protection assay revealed that the short variant of ovarian PACAPR was the predominant form stimulated during prepubertal development and by gonadotropins. These results demonstrate the expression of PACAPR messenger RNA in granulosa cells of growing follicles and of preovulatory follicles stimulated by gonadotropins, and suggest that PACAP may play a role in the growth of developing follicles and in ovulation as an autocrine/paracrine factor.


Subject(s)
Gene Expression Regulation/physiology , Ovarian Follicle/physiology , Ovary/physiology , Receptors, Pituitary Hormone/genetics , Transcription, Genetic , Aging , Animals , Cells, Cultured , Chorionic Gonadotropin/pharmacology , Female , Gene Expression Regulation/drug effects , Gonadotropins, Equine/pharmacology , Humans , Luteinizing Hormone/pharmacology , Ovarian Follicle/cytology , Ovarian Follicle/drug effects , Ovary/drug effects , Ovary/growth & development , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Sexual Maturation
6.
Endocrinology ; 140(2): 818-26, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9927311

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP), a novel neuropeptide with considerable homology to vasoactive intestinal peptide and GH-releasing hormone, exists in two biologically active forms, PACAP-38 and -27. The presence of PACAP in the ovary has been demonstrated, where it stimulates steroidogenesis and cAMP accumulation in cultured granulosa cells. In the present study, gonadotropin regulation of PACAP gene expression was examined in PMSG/human (h)CG-treated immature rat ovaries and cultured preovulatory follicles. Northern blot analysis of ovaries obtained from PMSG/hCG-treated immature animals revealed the transient induction of PACAP transcripts by hCG, reaching a maximum at 6 h. The major cell types expressing PACAP messenger RNA were granulosa cells of preovulatory follicles and some theca/interstitial cells. In preovulatory follicles cultured in serum-free medium, PACAP transcripts were transiently induced by LH and FSH, reaching a maximum 6-9 h after stimulation in granulosa cells but not in theca cells. Treatment with cycloheximide or alpha-amanitin abolished LH-induced PACAP transcripts, indicating that new protein synthesis and transcription are necessary. Treatment with MDL-12,330A, an inhibitor of adenylate cyclase, inhibited LH-induced PACAP messenger RNA, and forskolin mimicked the LH action, implying the role of adenylate cyclase activation. In contrast, treatment with chelerythrine, an inhibitor of protein kinase C, and 2-O-tetradecanol-phorbol-13-acetate had no effect. We further tested the role of PACAP in follicle apoptosis using apoptotic DNA fragmentation analysis. Treatment with PACAP-38 suppressed follicle apoptosis in a dose-dependent manner. Moreover, the LH suppression of follicle apoptosis was partially blocked by cotreatment with PACAP-38 antagonist, indicating mediation by endogenous PACAP-38. These results suggest that PACAP, transiently induced by the gonadotropin surge, could be a local regulator of a number of events and may act as a follicle survival factor during the periovulatory period.


Subject(s)
Gonadotropins/pharmacology , Neuropeptides/genetics , Ovarian Follicle/physiology , Ovary/metabolism , RNA, Messenger/metabolism , Adenylyl Cyclases/metabolism , Amanitins/pharmacology , Animals , Apoptosis/drug effects , Chorionic Gonadotropin/pharmacology , Cycloheximide/pharmacology , Enzyme Activation/physiology , Female , Follicular Phase/physiology , Luteinizing Hormone/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Ovary/drug effects , Pituitary Adenylate Cyclase-Activating Polypeptide , Protein Synthesis Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Sheep
7.
Endocrinology ; 142(9): 4015-25, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11517181

ABSTRACT

Recently we characterized three distinct GnRH receptors in the bullfrog (bfGnRHR-1, bfGnRHR-2, and bfGnRHR-3). In the present study, we further investigated the expression and function of splice variants, generated from the primary bfGnRHR-3 transcript by exon skipping (splice variant 1), intron retention (splice variants 2 and 3), and/or transcriptional slippage (splice variant 4), apart from the constitutively spliced form (wild-type). Cellular expression and function of the splice variants were examined using a transient expression system. Immunoblot analysis revealed that the wild-type receptor and all splice variant proteins were expressed in transfected HeLa cells with no significant differences in expression levels. These splice variants showed a very low binding affinity to ligand and did not induce signal transduction in response to GnRH treatment. Interestingly, cotransfection of the wild-type with splice variants 2--4, but not with splice variant 1, significantly inhibited wild-type receptor-mediated signaling. Subcellular localization analysis of green fluorescent protein-tagged wild-type and splice variant proteins revealed that the wild-type receptor protein was mainly localized in the cell membrane, whereas the splice variant 1 protein was exclusively detected in the cytoplasm. The splice variant 2--4 proteins, however, were found in both the cell membrane and cytoplasm. The inhibition of wild-type receptor signaling by splice variants 2--4 and the subcellular localization of splice variants 2-4 suggest a possible physical interaction of splice variants 2--4 with the wild-type receptor protein. In addition, the ratio of mRNA levels of the wild-type to splice variants 2--4 significantly varied from hibernation (wild-type < splice variants 2--4) to the prebreeding season (wild-type > splice variants 2--4). Collectively, these results suggest that alternative splicing of the bfGnRHR-3 primary transcript plays a role in fine-tuning GnRH receptor function in amphibians.


Subject(s)
Alternative Splicing , Genetic Variation/physiology , Rana catesbeiana/physiology , Receptors, LHRH/physiology , Signal Transduction/physiology , Amino Acid Sequence/genetics , Animals , Base Sequence/genetics , Biological Transport/physiology , Cell Membrane/metabolism , Gene Expression , Hibernation/physiology , Ligands , Molecular Sequence Data , RNA/metabolism , Rana catesbeiana/genetics , Receptors, LHRH/genetics , Reference Values , Reproduction/physiology , Seasons , Transcription, Genetic/physiology
8.
J Mol Endocrinol ; 27(3): 329-38, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11719285

ABSTRACT

The aim of our in vitro experiments was to examine if IGF binding protein (IGFBP)-3 is involved in control of bovine ovarian secretory activity. For this purpose we performed the transfection of bovine granulosa cells with cDNA sense and antisense constructs increasing or inhibiting IGFBP-3 synthesis. The release of IGFBP-3, progesterone, oxytocin, IGF-I and prostaglandins F (PGF) and E (PGE) by control and transfected cells was compared. The transfected ovarian cells were cultured with and without bLH (100 ng/ml), bGH (100 ng/ml), IGF-I (10 ng/ml), oxytocin (10 ng/ml) and oestradiol-17beta (100 ng/ml). The concentration of IGFBP-3 produced was assessed using ligand and western blotting and secretion of progesterone, oxytocin, IGF-I, PGF and PGE was evaluated using RIA/IRMA techniques. Transfection of cells with the sense IGFBP-3 cDNA construct resulted in the expected increase in IGFBP-3 release, whereas the antisense IGFBP-3 construct induced the expected reduction in IGFBP-3 output. The granulosa cells transfected to overexpress IGFBP-3 had an increase in IGF-I, PGF and PGE release, and a decrease in basal and hormone- or growth factor-induced accumulation of progesterone and oxytocin. The granulosa cells transfected to have reduced IGFBP-3 expression gave primarily significant opposite findings. The present results suggest the involvement of IGFBP-3 in control of bovine ovarian steroid, peptide hormone, growth factor and prostaglandin release. IGFBP-3 is a physiological stimulator of IGF-I and prostaglandin release and an inhibitor of steroid and peptide hormone output.


Subject(s)
Granulosa Cells/drug effects , Granulosa Cells/metabolism , Hormones/pharmacology , Insulin-Like Growth Factor Binding Protein 3/genetics , Animals , Cattle , DNA, Antisense/genetics , DNA, Complementary/genetics , Estradiol/pharmacology , Female , Growth Hormone/pharmacology , In Vitro Techniques , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Luteinizing Hormone/pharmacology , Oxytocin/metabolism , Oxytocin/pharmacology , Progesterone/metabolism , Prostaglandins E/metabolism , Prostaglandins F/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
9.
J Mol Endocrinol ; 33(1): 293-313, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15291760

ABSTRACT

The neurohypophysial nonapeptides vasotocin (VT) and mesotocin (MT) are the amphibian counterparts of arginine vasopressin (AVP) and oxytocin (OT). We have here reported the cloning and functional characterization of the receptors for vasotocin (VTR) and mesotocin (MTR) in two species of frog, Rana catesbeiana and Rana esculenta. The frog VTR and MTR cDNAs encode proteins of 419 and 384 amino acids respectively. Frog VTR exhibits a high degree of sequence identity with the mammalian AVP-1a (V1a) receptor while the frog MTR possesses a high degree of sequence identity with the mammalian OT receptor. Activation of VTR induced both c-fos promoter- and cAMP-responsive element (CRE)-driven transcriptional activities, while activation of MTR induced c-fos promoter-driven transcriptional activity but failed to evoke CRE-driven transcriptional activity, suggesting differential G protein coupling between VTR and MTR. The VTR exhibited the highest sensitivity for VT followed by OT>AVP approximately MT, whereas the MTR showed preferential ligand sensitivity for MT>OT>VT>AVP. A V1a agonist but not V2 and OT agonists substantially activated both VTR and MTR with a similar sensitivity. V1a, V2 and OT antagonists inhibited MT-induced MTR activation but not VT-induced VTR activation. In the frog brain, VTR and MTR mRNAs were found to be widely expressed in the telencephalon, diencephalon and mesencephalon, and exhibited very similar regional distribution. In the pituitary, VTR and MTR were expressed in the distal and intermediate lobes but were virtually absent in the neural lobe. Taken together, these data indicated that, although the distribution of VTR and MTR largely overlaps in the frog brain and pituitary, VT and MT may play distinct activities owing to the ligand selectivity and different signaling pathways activated by their receptors.


Subject(s)
Receptors, Pituitary Hormone/metabolism , Receptors, Vasopressin/metabolism , Amino Acid Sequence , Animals , Base Sequence , Brain/metabolism , Cloning, Molecular , DNA Primers , DNA, Complementary , Female , GTP-Binding Proteins/metabolism , Humans , In Situ Hybridization , Ligands , Male , Molecular Sequence Data , Pituitary Gland/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rana esculenta , Receptors, Pituitary Hormone/genetics , Receptors, Vasopressin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Signal Transduction
10.
J Endocrinol ; 159(2): 313-21, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9795373

ABSTRACT

The aim of our in vitro experiments with isolated porcine ovarian follicles was to study the effects of gonadotropins, GH, IGF-I and oxytocin (OT) on release of ovarian steroid, OT, IGF-I, insulin-like growth factor-binding protein-3 (IGFBP-3), prostaglandin F (PGF), prostaglandin E (PGE) and cAMP. It was found that quarters of ovarian follicles cultured for 8 days produced significant amounts of progesterone, estradiol-17 beta, OT and IGFBP-3 with peaks of accumulation from the 3rd to the 8th day of culture. Addition of serum promoted progesterone, estradiol and OT release, whilst accumulation of IGFBP-3 was maintained to a greater extent in serum-free medium. GH (10 ng/ml or above) was able to inhibit androstenedione, OT, PGF and IGFBP-3, to stimulate IGF-I and cAMP, and to alter testosterone and PGE release by follicles cultured in serum-supplemented and/or serum-free medium. IGF-I (10 ng/ml or more) inhibited androstenedione and PGF secretion, stimulated testosterone, estradiol, OT and cAMP production, but did not influence progesterone, IGFBP-3 or PGE output in these conditions. OT (100 ng/ml) was able to inhibit androstenedione and to stimulate testosterone, IGF-I, PGF and PGE, but not estradiol or IGFBP-3 release. A stimulatory effect of LH on progesterone and OT and an inhibitory influence of LH on estradiol secretion in the serum-supplemented medium were observed. FSH in these conditions stimulated OT, but not progesterone or estradiol secretion. The use of this experimental model suggests the involvement of gonadotropins, OT, GH and IGF-I in the control of ovarian steroid and nonapeptide hormone, growth factor, growth factor-binding protein, prostaglandin and cyclic nucleotide production. The stimulatory effect of GH on IGF-I, and the stimulatory influence of IGF-I on OT, as well as coincidence of the majority of effects of IGF-I and OT, suggest the existence of a GH-IGF-I-OT axis. On the other hand, the different patterns of action of GH and IGF-I on OT, estrogen and IGFBP-3 suggest that part of the GH effect on ovarian cells is IGF-I independent.


Subject(s)
Gonadotropins, Pituitary/pharmacology , Growth Hormone/pharmacology , Ovarian Follicle/metabolism , Androstenedione/metabolism , Animals , Culture Media , Cyclic AMP/metabolism , Estradiol/metabolism , Female , Follicle Stimulating Hormone/pharmacology , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/metabolism , Luteinizing Hormone/pharmacology , Models, Biological , Ovarian Follicle/drug effects , Oxytocin/metabolism , Progesterone/metabolism , Prostaglandins E/metabolism , Prostaglandins F/metabolism , Radioimmunoassay , Recombinant Proteins/pharmacology , Swine , Testosterone/metabolism
11.
J Endocrinol ; 171(3): 475-80, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11739013

ABSTRACT

The aims of this study on porcine ovarian granulosa cells were to examine the effect of GH on oxytocin (OT), IGF-I and IGF-I receptors, IGF-binding protein-3 (IGFBP-3), progesterone and prostaglandin E (PGE), as well as to determine whether IGF-I and/or OT may be mediators of GH action. The cells were cultured either with porcine GH (pGH) (1 ng/ml to 10 microg/ml or 100 ng/ml only), antiserum against IGF-I (0.1%), antiserum against OT (0.1%) or a combination of GH (10 ng/ml) with antiserum against IGF-I or antiserum against OT (0.1%). The secretion of IGF-I, OT, IGFBP-3, progesterone and PGE was determined using RIA/IRMA, whilst the IGF-I binding sites were measured using a radioreceptor assay. It was observed that pGH increased the secretion of IGF-I and the abundance of IGF-I binding sites in granulosa cells. Furthermore, GH inhibited OT release, stimulated progesterone and PGE output, but had no significant effect on IGFBP-3 secretion. Immunoneutralization of IGF-I by antiserum against IGF-I inhibited PGE secretion, but it did not influence progesterone or IGFBP-3 secretion. Binding of OT by antiserum suppressed IGFBP-3, PGE, but not progesterone secretion. Neither immunoneutralization of IGF-I nor OT substantially prevented the effects of GH on progesterone, IGFBP and PGE. These observations demonstrate the involvement of GH, IGF-I and OT in the control of porcine ovarian secretory activity and the ability of GH to regulate IGF-I and OT production and IGF-I reception. Nevertheless, lack of correlation between the effects of GH, antiserum against IGF-I and antiserum against OT, as well as the inability of blockade of IGF-I or OT to prevent the effects of GH, suggests that IGF-I and OT, despite their dependence on GH, do not mediate GH action on ovarian cells.


Subject(s)
Granulosa Cells/drug effects , Growth Hormone/pharmacology , Swine/metabolism , Animals , Cell Culture Techniques , Culture Media, Conditioned , Female , Granulosa Cells/metabolism , Growth Hormone/physiology , Immune Sera , Insulin-Like Growth Factor Binding Protein 3/biosynthesis , Insulin-Like Growth Factor I/antagonists & inhibitors , Insulin-Like Growth Factor I/biosynthesis , Insulin-Like Growth Factor I/physiology , Oxytocin/antagonists & inhibitors , Oxytocin/biosynthesis , Oxytocin/physiology , Progesterone/biosynthesis , Prostaglandins E/biosynthesis
12.
Mol Cell Endocrinol ; 205(1-2): 193-9, 2003 Jul 31.
Article in English | MEDLINE | ID: mdl-12890581

ABSTRACT

Growth hormone (GH), prostaglandins F (PGF) and prostaglandins E (PGE) are important regulators of ovarian function. Therefore, interrelationships between GH and these substances and their intracellular mechanisms might be of physiological significance in the ovary. The aims of this study on cultured porcine ovarian granulosa cells were to determine the effect of GH on the secretion of oxytocin (OT), PGF and PGE and whether MAP kinase could be involved in the mediation of GH action. Experiments were carried out with cultured porcine granulosa cells to investigate the effects of exogenous pGH (1-100 ng/ml) on the expression of MAP kinase (ERK-1, -2) and of PGH (1-100 ng/ml) and the MAP kinase blocker PD 98059 (1 microg/ml) on the secretion of PGF, PGE and OT. The cellular content of ERK-1 and -2 was analyzed by Western immunoblotting and immunocytochemistry, whilst PGF, PGE and OT accumulation in the medium was measured by RIA. Addition of GH to culture medium significantly altered the pattern of ovarian ERK MAP kinase on SDS-PA gels: the 44 and 42 kDa bands were reduced and additional 50 and 48 kDa bands appeared. Moreover, there was an increase in the percentage of cells containing ERK MAP kinase. GH stimulated the secretion of PGF (at a concentration of 1 ng GH per ml medium) and OT (100 ng GH per ml), but not PGE. The MAP kinase blocker alone did not affect PGF, PGE and OT secretion but did prevent the stimulatory effects of GH on PGF and induced stimulatory action of GH (10 ng/ml) on PGE. GH-stimulated OT secretion was unaffected. These observations confirm the role of GH in regulating porcine ovarian PGF, PGE and OT secretion and the presence of ERK MAP kinase in porcine granulosa cells. Furthermore, our studies demonstrate that MAP kinase-dependent intracellular mechanisms are dependent on GH, and that these mechanisms are involved in the mediation of GH action on ovarian PGF and PGE but not OT secretion.


Subject(s)
Granulosa Cells/enzymology , Growth Hormone/pharmacology , Mitogen-Activated Protein Kinases/physiology , Animals , Cells, Cultured , Female , Granulosa Cells/drug effects , Granulosa Cells/physiology , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Signaling System , Oxytocin/biosynthesis , Prostaglandins/biosynthesis , Swine
13.
Mol Cell Endocrinol ; 148(1-2): 1-10, 1999 Feb 25.
Article in English | MEDLINE | ID: mdl-10221765

ABSTRACT

The steroidogenic acute regulatory (StAR) protein plays essential roles in the delivery of cytosolic cholesterol into the mitochondrial inner membrane, which is an acute regulated and rate-limiting step for the steroid hormone synthesis. Since retinoic acids (RAs) are known to induce the synthesis of steroid hormones in mouse Leydig cells in vitro, mouse Leydig tumour cells, K28, were used to determine the effect of RAs on the level of StAR mRNA by Northern blot analysis. The level of StAR mRNA reached the maximum in a 4-8 h treatment with all-trans-RA (atRA) or 9-cis-RA (9cRA), and the effects were dose-dependent. The effect of 9cRA on the levels of StAR mRNA was blocked by actinomycin D, which indicates that 9cRA might exert a direct effect on the transcription of the gene. Promoter/reporter constructs containing a 5'-flanking region of the mouse or rat StAR gene were prepared, and luciferase activity was assayed following transient transfection into K28 or adrenal tumour cells, Y1. The result revealed that the luciferase activity was increased by 4-5-fold in response to the treatment of 9cRA, which indicated that 9cRA participates transcriptional activation of the StAR protein gene.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Phosphoproteins/genetics , Tretinoin/pharmacology , Alitretinoin , Animals , Bucladesine/pharmacology , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Kinetics , Leydig Cell Tumor , Mice , Mutagenesis, Site-Directed , Neoplasm Proteins/genetics , Phosphoproteins/physiology , Progesterone/biosynthesis , RNA, Messenger/genetics , Rats , Sequence Deletion , Transcription, Genetic/drug effects , Tumor Cells, Cultured
14.
Mol Cell Endocrinol ; 164(1-2): 197-204, 2000 Jun.
Article in English | MEDLINE | ID: mdl-11026571

ABSTRACT

To date nine structural variants of GnRH have been identified in vertebrates and two additional forms have been isolated from a tunicate. In amphibians only mammalian GnRH ([Arg8] GnRH) and type II GnRH (chicken GnRH II, [His5, Trp7, Tyr8] GnRH) have been identified. In the present study, a full-length cDNA encoding a novel type of GnRH was isolated from pituitary of Rana dybowskii. The GnRH gene encodes a GnRH peptide ([Trp8] GnRH) in which tryptophan is substituted for arginine of mammalian GnRH Northern blot analysis revealed the presence of a single 500 bp transcript for the [Trp8] GnRH precursor in forebrain but its absence in testis, ovary, kidney and liver. Restriction digests of genomic DNA demonstrated a single copy of the gene. The [Trp8] GnRH immunoreactive cells were identified in the preoptic area of the frog brain. Synthetic [Trp8] GnRH was tested for its ability to stimulate inositol phosphate production by COS-1 cells transfected with the cloned Xenopus pituitary GnRH receptor and the cloned human GnRH receptor. [Trp8] GnRH had a potency of about 60% compared with mammalian GnRH ([Arg8] GnRH) for the Xenopus receptor, whereas the potency of [Trp8] GnRH was approximately 5% compared with mammalian GnRH for the human receptor. Both mammalian GnRH and [Trp8] GnRH were 1000-fold less potent than type II GnRH for the Xenopus GnRH receptor. The similar potency of [Arg8] GnRH and the novel [Trp8] GnRH for the Xenopus pituitary receptor indicates that, unlike the human receptor, the Xenopus receptor does not discriminate between these amino acids in position eight thereby allowing substitution of the arginine in the mammalian GnRH.


Subject(s)
Brain/metabolism , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/analysis , DNA, Complementary/genetics , Gonadotropin-Releasing Hormone/analysis , Gonadotropin-Releasing Hormone/pharmacology , Humans , Immunohistochemistry , Molecular Sequence Data , Ranidae , Sequence Alignment
15.
Brain Res Mol Brain Res ; 50(1-2): 51-8, 1997 Oct 15.
Article in English | MEDLINE | ID: mdl-9406917

ABSTRACT

To examine whether an ultrashort feedback mechanism of gonadotropin-releasing hormone (GnRH) operates at the level of gene transcription, we studied the effects of GnRH analogs on GnRH promoter activity and GnRH mRNA level in hypothalamic GT1-1 neuronal cells. Treatment of GT1-1 cells with buserelin, a GnRH agonist, or native GnRH for 24 h significantly decreased GnRH promoter activity and its mRNA level, whereas that with GnRH antagonists, antide or [D-Phe2,D-Ala6]-GnRH, showed no effect. The inhibitory effects of buserelin on GnRH gene transcription and GnRH mRNA level were dose-related, and a significant inhibition was observed in cells treated with buserelin at concentrations higher than 0.1 microM. Time-course experiments showed that significant decreases in GnRH promoter-driven luciferase activity and GnRH mRNA level were observed within 12 h and sustained up to 48 h. Moreover, treatment with GnRH agonist for 12 h significantly decreased the transcription rate of the mouse GnRH gene, as revealed by nuclear run-on transcription assay. The promoter analysis with the 5'-deletional constructs demonstrated that cis-acting elements important for GnRH autoregulation by GnRH agonist reside within -854 bp upstream from the transcription start site. These data clearly demonstrate that GnRH can exert autocrine regulation at the level of GnRH gene transcription.


Subject(s)
Autocrine Communication , Gonadotropin-Releasing Hormone/genetics , Hypothalamus/metabolism , Neurons/metabolism , Transcription, Genetic , Animals , Buserelin/pharmacology , Cell Count , Cell Line , Feedback , Hypothalamus/cytology , Hypothalamus/drug effects , Mice , Neurons/drug effects
16.
Brain Res Mol Brain Res ; 53(1-2): 226-35, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9473680

ABSTRACT

The present study is designed to evaluate the relationship between gonadotropin-releasing hormone (GnRH) and GnRH receptor (GnRHR) gene expression during the steroid-induced LH surge. One week after ovariectomy (OVX), a capsule containing 17beta-estradiol (E) or vehicle (V) was implanted into OVX rats, and 2 days later a single injection of progesterone (P) or V was administered s.c. at 10:00 h. Poly(A)-rich RNA samples were isolated from the micropunches of the preoptic area (POA) and the posterior mediobasal hypothalamus (pMBH) from both sides of individual brain slices. Using competitive reverse transcription-polymerase chain reaction (RT-PCR) procedures, three parameters (POA GnRH, pMBH GnRHR) and pituitary GnRHR mRNA levels were simultaneously determined in each individual animal. POA GnRH mRNA and pituitary GnRHR mRNA levels were decreased by treatment with E, but increased by a combination of E and P. In contrast, pMBH GnRHR mRNA levels were clearly augmented by treatment with E, and decreased by the combination of E and P. Temporal changes in such parameters were determined in OVX+E+V- and OVX+E+P-treated rats. P augmented POA GnRH mRNA levels at the time of the LH surge (17:00 h) and the increased GnRH mRNA levels were remained until 22:00 h, while E alone failed to alter POA GnRH mRNA levels. In the pMBH micropunch samples, P substantially decreased E-induced increase in GnRHR mRNA levels at 17:00 h and further lowered those until 22:00 h. Antisense oligonucleotides of GnRHR mRNA administered into the lateral ventricle of OVX+E-treated rats blocked the E-induced increase in pMBH GnRHR mRNA levels. The antisense oligonucleotides also prevented the LH surge as well as the increase in pituitary GnRHR mRNA levels in the OVX+E+P-treated group. However, administration of this antisense oligonucleotides failed to alter POA GnRH mRNA levels. In conclusion, the present study demonstrated that there is an inverse relationship between POA GnRH mRNA levels and pMBH GnRHR mRNA levels in response to E and/or P, and that the blockade of the E-induced increase in pMBH GnRHR mRNA levels effectively nullified the P-induced LH surge. These results indicate that pMBH GnRHR gene expression is involved in synchronizing the GnRH neuronal activity, which is crucial for the generation of the LH surge.


Subject(s)
Estradiol/pharmacology , Gene Expression Regulation/drug effects , Hypothalamus, Middle/physiology , Luteinizing Hormone/blood , Neurons/physiology , Progesterone/pharmacology , Receptors, LHRH/biosynthesis , Animals , DNA Primers , Drug Implants , Estradiol/administration & dosage , Female , Hypothalamus, Middle/drug effects , Neurons/drug effects , Oligonucleotides, Antisense/pharmacology , Ovariectomy , Pituitary Gland/physiology , Polymerase Chain Reaction , Preoptic Area/drug effects , Preoptic Area/physiology , Rats , Rats, Sprague-Dawley , Transcription, Genetic/drug effects
17.
Brain Res Mol Brain Res ; 78(1-2): 59-68, 2000 May 31.
Article in English | MEDLINE | ID: mdl-10891585

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) regulates pituitary hormone biosynthesis and secretion through its cognate receptors. PACAP also plays an important role in the regulation of ovarian steroid biosynthesis. If so, there might be a feedback regulation of hypothalamic PACAP synthesis by the pituitary and by ovarian steroids. In the present study, we used RNase protection assays to determine changes in mRNA levels of PACAP and type I PACAP receptor (PAC(1)) under the conditions of ovariectomy and replacement with ovarian steroids. Progesterone (P) alone or in combination with estradiol (E) induced significant increases in PACAP mRNA level in the medial basal hypothalamus (MBH) and PAC(1) mRNA levels in MBH and the preoptic area (POA). This finding suggests that feedback regulation takes place between the ovary and hypothalamic PACAP neurons. P is known to be a major regulatory feedback factor for hypothalamic luteinizing hormone-releasing hormone (LHRH) neurons, but P receptor is not present in these neurons. Therefore, we examined a possible involvement of PACAP in the feedback regulatory pathway of P to LHRH neurons. After an antisense PAC(1) oligodeoxynucleotide (ODN) was i.c.v.-injected into the third ventricle of E and P-treated rats, LHRH mRNA levels were determined. The ODN markedly decreased the P-induced increase in the LHRH mRNA level. Taken together, the present data suggest that PACAP may play a role as a mediator in the regulation of LHRH synthetic machinery by stimulatory feedback of P.


Subject(s)
Hypothalamus, Middle/physiology , Neuropeptides/genetics , Pituitary Gland, Anterior/physiology , Progesterone/pharmacology , Receptors, Pituitary Hormone/genetics , Animals , Antisense Elements (Genetics) , Brain Chemistry/drug effects , Brain Chemistry/genetics , Feedback/physiology , Female , Gene Expression/drug effects , Gene Expression/physiology , Gonadotropin-Releasing Hormone/genetics , Hypothalamus, Middle/cytology , Injections, Intraventricular , Neurons/chemistry , Neurons/physiology , Ovariectomy , Pituitary Adenylate Cyclase-Activating Polypeptide , Pituitary Gland, Anterior/cytology , Preoptic Area/cytology , Preoptic Area/physiology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide
18.
J Neuroendocrinol ; 10(12): 911-8, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9870748

ABSTRACT

The present study was designed to investigate whether noradrenergic neurotransmission regulates the gene expression of gonadotropin-releasing hormone (GnRH) in the preoptic area and GnRH receptor in the pituitary. To this end, N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP4, 50 mg/kg), an intraperitoneal (i.p.) injection of selective noradrenergic neurotoxin, was administered 1 h before progesterone (1 mg) treatment in ovariectomized and estradiol-treated prepubertal rats. Treatment with DSP4 effectively blocked the progesterone-induced increase in hypothalamic noradrenaline content, but not dopamine content, indicating that DSP4 selectively inhibits noradrenergic neurotransmission. DSP4 significantly blocked progesterone-induced increase in serum luteinizing hormone (LH) concentrations as well as GnRH release from hypothalamic fragments incubated in vitro. DSP4 concomitantly down-regulated GnRH mRNA levels in the preoptic area, as determined by competitive reverse transcription-polymerase chain reaction. DSP4 also clearly down-regulated progesterone-induced GnRH receptor mRNA levels in the pituitary, whereas it failed to alter LHbeta mRNA levels. In summary, blockade of noradrenergic neurotransmission with DSP4 resulted in profound reductions of hypothalamic GnRH and pituitary GnRH receptor gene expression.


Subject(s)
Adrenergic Agents/pharmacology , Benzylamines/pharmacology , Down-Regulation/drug effects , Gonadotropin-Releasing Hormone/genetics , Receptors, LHRH/genetics , Steroids/pharmacology , Animals , Benzylamines/administration & dosage , Dopamine/metabolism , Estradiol/pharmacology , Female , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Injections, Intraperitoneal , Luteinizing Hormone/blood , Luteinizing Hormone/genetics , Neurotoxins/pharmacology , Norepinephrine/metabolism , Ovariectomy , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Progesterone/administration & dosage , Progesterone/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
19.
Mol Cells ; 7(4): 526-31, 1997 Aug 31.
Article in English | MEDLINE | ID: mdl-9339898

ABSTRACT

Yeast artificial chromosome (YAC) clones have been successfully utilized to generate a YAC contig map of the long arm of human chromosome 21 (Hu21q). The chromosome subband of 21q22.1 where five genetic loci (IFNAR1, IFNAR2, CRFB4, AF-1, and GART) are mapped is a gene-rich region and needs to be characterized in further detail. YAC D142H8 and YAC F136C5, which were characterized previously by a functional YAC expression procedure, and two new YAC clones, YAC 872B5 and YAC 876D4 located at 21q22.1 whose YAC sizes are 800 kb and 1,500 kb, respectively, were used in this study. To obtain more markers useful for making a detailed physical map of the region, a purified yeast artificial chromosome (YAC D142H8) was used to screen the 3 x 1 S cDNA library. As a result three anonymous cDNA clones (Kmy1, Kmy2, and Qorf4) were obtained, and the nucleotide sequences of Kmy1 and Kmy2 were determined. In an attempt to make a detailed physical map of the region, the locations of five known genes as well as the three new markers were determined on the four YACs by Southern blot analysis. YAC 872B5 contained all markers except GART while YAC F136C5, YAC D142H8, and YAC 876D4 contained three markers (CRFB4, IFNAR1, and IFNAR2), four markers (Kmy1, Kmy2, Qorf4 and AF-1), and four markers (Kmy1, Kmy2, Qorf4 and GART), respectively. YAC 872B5 may represent 1,500 kb of the 21q22.1 subband and half of the 3 x 1 S region, so it should be very useful for studying the relevent region of the human chromosome functionally and physically.


Subject(s)
Chromosome Mapping , Chromosomes, Artificial, Yeast/chemistry , Chromosomes, Human, Pair 21/chemistry , Base Sequence , Blotting, Southern , Chromosomes, Artificial, Yeast/genetics , Chromosomes, Human, Pair 21/genetics , Cloning, Molecular , DNA, Complementary/analysis , Genetic Markers , Humans , Hybrid Cells , Molecular Sequence Data
20.
Mol Cells ; 10(3): 263-8, 2000 Jun 30.
Article in English | MEDLINE | ID: mdl-10901163

ABSTRACT

In yeast, trehalose-6-phosphate synthase is a key enzyme for trehalose biosynthesis, encoded by the structural gene TPS1. Trehalose affects sugar metabolism as well as osmoprotection against several environmental stresses, such as heat and desiccation. The TPS1 gene of Saccharomyces cerevisiae was engineered under the control of the CaMV 35S promoter for constitutive expression in transgenic potato plants by Ti-plasmid of Agrobacterium-mediated transformation. The resulting TPS1 transgenic potato plants exhibited various morphological phenotypes in culture tubes, ranging from normal to severely retarded growth, including dwarfish growth, yellowish lancet-shaped leaves, and aberrant root development. However, the plants recovered from these negative growth effects when grown in a soil mixture. The TPS1 transgenic potato plants showed significantly increased drought resistance. These results suggest that the production of trehalose not only affects plant development but also improves drought tolerance.


Subject(s)
Genetic Engineering , Glucosyltransferases/genetics , Saccharomyces cerevisiae/genetics , Solanum tuberosum/physiology , Agrobacterium tumefaciens/genetics , DNA, Plant/genetics , DNA, Plant/metabolism , Disasters , Glucosyltransferases/metabolism , Phenotype , Plants, Genetically Modified , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Saccharomyces cerevisiae/enzymology , Solanum tuberosum/genetics , Transformation, Genetic , Water
SELECTION OF CITATIONS
SEARCH DETAIL