Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 301
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Med ; 7(5): 605-11, 2001 May.
Article in English | MEDLINE | ID: mdl-11329063

ABSTRACT

Adaptive stress responses mediated by the endocrine, autonomic, cardiovascular and immune systems are essential for the survival of the individual. Initial stress-induced responses provide a vital short-term metabolic lift, but prolonged or inappropriate exposure to stress can compromise homeostasis thereby leading to disease. This 'fight-or-flight' response is characterized by the activation of the corticotropin-releasing hormone (CRH)-adrenocorticotropin-glucocorticoid axis, mediated by the type 1 CRH receptor. In contrast, the type 2 CRH receptor mediates the stress-coping responses during the recovery phase of stress. We identified human stresscopin (SCP) and stresscopin-related peptide (SRP) as specific ligands for the type 2 CRH receptor. The genes encoding these peptides were expressed in diverse peripheral tissues as well as in the central nervous system. Treatment with SCP or SRP suppressed food intake, delayed gastric emptying and decreased heat-induced edema. Thus SCP and SRP might represent endogenous ligands for maintaining homeostasis after stress, and could allow the design of drugs to ameliorate stress-related diseases.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Amino Acid Sequence , Animals , Base Sequence , Corticotropin-Releasing Hormone/chemistry , Corticotropin-Releasing Hormone/genetics , DNA, Complementary , Humans , Immunohistochemistry , Ligands , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Rats , Rats, Sprague-Dawley , Sequence Homology, Amino Acid , Urocortins
2.
Science ; 204(4395): 854-5, 1979 May 25.
Article in English | MEDLINE | ID: mdl-375393

ABSTRACT

Gonadotropin-releasing hormone (GnRH) and its agonistic analogs inhibited the follicle-stimulating hormone (FSH)-induced increase of estrogen and progesterone production in vitro by rat ovarian granulosa cells. Likewise, GnRH analogs inhibited FSH-induced changes in ovarian function in hypophysectomized rats in vivo. These results indicate that GnRH, in addition to its well-known gonadotropin-releasing action in the pituitary, exerts a direct inhibition of ovarian steroidogenesis.


Subject(s)
Estrogens/biosynthesis , Follicle Stimulating Hormone/antagonists & inhibitors , Gonadotropin-Releasing Hormone/pharmacology , Ovary/drug effects , Progestins/biosynthesis , Animals , Cells, Cultured , Female , Granulosa Cells/drug effects , Hypophysectomy , Ovary/metabolism , Rats
3.
Science ; 239(4835): 72-4, 1988 Jan 01.
Article in English | MEDLINE | ID: mdl-3122320

ABSTRACT

Follicle-stimulating hormone (FSH) is a glycoprotein essential for gonadal development and steroidogenesis. Recent studies suggest that deglycosylation of FSH results in the formation of antagonistic proteins that are capable of binding to gonadal receptors but that are devoid of bioactivity. Treatment of hypogonadal women with an antagonist of gonadotropin-releasing hormone substantially decreased serum FSH bioactivity with minimal changes in immunoreactivity. Chromatofocusing and size fractionation of the serum samples indicated the secretion of immunoreactive FSH isoforms that are devoid of bioactivity but that are capable of blocking FSH action in ovarian granulosa cells. These findings provide the first demonstration of naturally occurring circulating antihormones. These FSH antagonists may play an important role in the physiology and pathophysiology of the gonads.


Subject(s)
Follicle Stimulating Hormone/antagonists & inhibitors , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Biological Assay , Cross Reactions , Female , Follicle Stimulating Hormone/immunology , Follicle Stimulating Hormone/metabolism , Glycoproteins/physiology , Humans , Isoelectric Point , Radioligand Assay , Structure-Activity Relationship
4.
Endocr Rev ; 21(2): 200-14, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10782364

ABSTRACT

Mammalian ovaries consist of follicles as basic functional units. The total number of ovarian follicles is determined early in life, and the depletion of this pool leads to reproductive senescence. Each follicle develops to either ovulate or, more likely, to undergo degeneration. The dynamics of ovarian follicle development have interested endocrinologists and developmental biologists for many years. With the advent of assisted reproductive techniques in humans, the possibility of regulating follicle development in vivo and in vitro has gained clinical relevance. In this review, we focus upon key branching points during the development of ovarian follicles as well as factors involved in determining the eventual destiny of individual follicles. We discuss inconsistencies in the literature regarding the definitions of follicle recruitment and selection and propose to name the two major steps of follicle development as initial and cyclic recruitment, respectively. Because some of these disparities have arisen due to differences in the animal systems studied, we also compare the development of the ovarian follicles of both humans and rats. We also review the status of knowledge of several puzzling clinical issues that may provide important clues toward unlocking the mechanisms of follicle development.


Subject(s)
Ovarian Follicle/physiology , Aging , Animals , Female , Follicular Atresia , Humans , Ovarian Follicle/growth & development , Ovulation , Pregnancy , Reproduction/physiology
5.
J Clin Invest ; 102(3): 532-7, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9691090

ABSTRACT

During each reproductive cycle, a preovulatory surge of gonadotropins induces meiotic maturation of the oocyte in the preovulatory follicle followed by ovulation. Although gonadotropins stimulate cAMP production in somatic cells of the follicle, a decrease in intra-oocyte cAMP levels is required for resumption of meiosis in oocytes. Based on the observed compartmentalization of the cAMP-degrading enzyme, phosphodiesterase, in follicular somatic and germ cells, inhibitors of phosphodiesterase 3 were used to block meiosis in ovulating oocytes in rodents. By this strategy, we demonstrated that fertilization and pregnancy could be prevented without disturbing follicle rupture and normal estrous cyclicity. In contrast to conventional contraceptive pills that disrupt ovarian steroidogenesis and reproductive cycles, the present strategy achieves effective contraception by selective blockage of oocyte maturation and development without alterations in ovulation and reproductive cyclicity.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Contraceptive Agents, Female/pharmacology , Cyclic AMP/physiology , Estrus/drug effects , Meiosis/drug effects , Oogenesis/drug effects , Ovulation/drug effects , Phosphodiesterase Inhibitors/pharmacology , Second Messenger Systems/physiology , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Cyclic Nucleotide Phosphodiesterases, Type 3 , Female , Fertilization/drug effects , Heart Rate/drug effects , Hypoxanthine/pharmacology , Isoenzymes/antagonists & inhibitors , Menotropins/pharmacology , Mice , Mice, Inbred C57BL , Milrinone , Ovarian Follicle/drug effects , Ovarian Follicle/physiology , Ovulation Induction , Pregnancy , Purinones/pharmacology , Pyridazines/pharmacology , Pyridones/pharmacology , Pyrrolidinones/pharmacology , Quinolones/pharmacology , Rats , Rats, Sprague-Dawley , Rolipram , Substrate Specificity , Thiophenes/pharmacology
6.
Nat Biotechnol ; 15(7): 663-7, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9219270

ABSTRACT

Glycoprotein hormones are noncovalent heterodimers comprised of a common alpha subunit and a hormone-specific beta subunit. Secretion and biologic action of these hormones are dependent on the formation of the heterodimer. The human LH beta subunit is unique among the other beta subunits in that it assembles inefficiently with the alpha subunit. To bypass this rate-limiting step, we constructed the LH single chains where the carboxy terminus of beta was fused to the amino terminus of alpha subunit through a linker. Compared to the human LH heterodimer, the extent of secretion was greater for the tethers although the rate was dependent on the nature of the linker. The LH single chains were biologically active even though there was loss of recognition by a LH-specific monoclonal antibody. This suggests that receptor binding of the single chains is not impaired by changes in the heterodimeric configuration resulting from tethering the subunits. In addition, single chains exhibited a remarkably greater in vitro stability than the heterodimer, implying that these analogs will be useful as diagnostic reagents and that their purification will be facilitated.


Subject(s)
Luteinizing Hormone/analogs & derivatives , Animals , Base Sequence , Biotechnology , CHO Cells , Cricetinae , DNA Primers/genetics , Drug Design , Drug Stability , Humans , In Vitro Techniques , Luteinizing Hormone/genetics , Luteinizing Hormone/pharmacology , Protein Conformation , Protein Engineering , Receptors, LH/metabolism , Transfection
7.
J Endocrinol ; 189(2): 329-39, 2006 May.
Article in English | MEDLINE | ID: mdl-16648300

ABSTRACT

In addition to gonadotropins, steroidogenesis and proliferation of granulosa cells during follicular development are controlled by a number of intraovarian factors including growth differentiation factor-9 (GDF-9), bone morphogenetic protein-4 (BMP-4), and IGF-I. The objective of this study was to determine the effect of GDF-9 and BMP-4 and their interaction with IGF-I and FSH on ovarian granulosa cell function in cattle. Granulosa cells from small (1-5 mm) and large (8-22 mm) follicles were collected from bovine ovaries and cultured for 48 h in medium containing 10% fetal calf serum and then treated with various hormones in serum-free medium for an additional 48 h. We evaluated the effects of GDF-9 (150-600 ng/ml) and BMP-4 (30 ng/ml) during a 2-day exposure on hormone-induced steroidogenesis and cell proliferation. In FSH plus IGF-I-treated granulosa cells obtained from small follicles, 300 ng/ml GDF-9 reduced (P < 0.05) progesterone production by 15% and 600 ng/ml GDF-9 completely blocked (P < 0.01) the IGF-I-induced increase in progesterone production. In comparison, 300 and 600 ng/ml GDF-9 decreased (P < 0.05) estradiol production by 27% and 71% respectively, whereas 150 ng/ml GDF-9 was without effect (P > 0.10). Treatment with 600 ng/ml GDF-9 increased (P < 0.05) numbers (by 28%) of granulosa cells from small follicles. In the same cells treated with FSH but not IGF-I, co-treatment with 600 ng/ml GDF-9 decreased (P < 0.05) progesterone production (by 28%), increased (P < 0.05) cell numbers (by 60%), and had no effect (P > 0.10) on estradiol production. In FSH plus IGF-I-treated granulosa cells obtained from large follicles, GDF-9 caused a dose-dependent decrease (P<0.05) in IGF-I-induced progesterone (by 13-48%) and estradiol (by 20-51%) production. In contrast, GDF-9 increased basal and IGF-I-induced granulosa cell numbers by over 2-fold. Furthermore, treatment with BMP-4 also inhibited (P < 0.05) steroidogenesis by 27-42% but had no effect on cell numbers. To elucidate downstream signaling pathways, granulosa cells from small follicles were transfected with similar to mothers against decapentaplegics (Smad) binding element (CAGA)- or BMP response element (BRE)-promoter reporter constructs. Treatment with GDF-9 (but not BMP-4) activated the Smad3-induced CAGA promoter activity, whereas BMP-4 (but not GDF-9) activated the Smad1/5/8-induced BRE promoter activity. We have concluded that bovine granulosa cells are targets of both GDF-9 and BMP-4, and that oocyte-derived GDF-9 may simultaneously promote granulosa cell proliferation and prevent premature differentiation of the granulosa cells during growth of follicles, whereas theca-derived BMP-4 may also prevent premature follicular differentiation.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , Follicle Stimulating Hormone/physiology , Granulosa Cells/physiology , Insulin-Like Growth Factor I/physiology , Intercellular Signaling Peptides and Proteins/pharmacology , Animals , Bone Morphogenetic Protein 4 , Cattle , Cell Count , Cell Division/physiology , Cell Line , Culture Media, Conditioned , Dose-Response Relationship, Drug , Estradiol/biosynthesis , Female , Granulosa Cells/drug effects , Growth Differentiation Factor 9 , Progesterone/biosynthesis , Promoter Regions, Genetic/genetics , Recombinant Proteins/pharmacology , Signal Transduction/physiology , Smad Proteins/metabolism
8.
Cancer Res ; 41(5): 1646-51, 1981 May.
Article in English | MEDLINE | ID: mdl-6260347

ABSTRACT

In an investigation comparing two strains of mice (BALB/c, susceptible to estrogen-induced Leydig cell tumors and C3H, resistant to such tumors), we found that the Leydig cell-luteinizing hormone (LH) receptors increase in BALB/c mice and decrease in C3H mice during estrogen treatment. In the BALB/c strain, LH receptor content in the tested of mice treated 1, 2, 4, 6, or 8 weeks with diethylstilbestrol (DES) was 2.4- to 5.4-fold greater than that in the testes of untreated littermates. By 24 weeks of treatment, the receptor number had increased 10-fold. Likewise, two weeks of estradiol benzoate treatment in BALB/c mice resulted in a dose-dependent increase in LH receptor content. In contrast, in C3H mice, DES treatment resulted in a transient initial increase (60%), followed by a time-dependent decrease in testicular LH receptor number: 38 and 17% that of normal by six and eight weeks of treatment, respectively. In both strains of mice, DES-induced changes in 125I-labeled human chorionic gonadotropin binding reflected changes in LH receptor number rather than in receptor affinity (approximately 3 x 10(-11) M). The testis weights of BALB/c mice remained normal during DES treatment, whereas those of the C3H decreased with time. Sprague-Dawley rats, resistant to estrogen-induced Leydig cell tumors, like C3H mice, also underwent testicular atrophy and lost LH receptors during DES treatment. The present study demonstrates that estrogen treatment indices diametrically oppossed change in testicular LH receptor number in the two strains of mice with different susceptibilities to Leydig cell tumorigenesis.


Subject(s)
Diethylstilbestrol/pharmacology , Leydig Cell Tumor/metabolism , Precancerous Conditions/metabolism , Receptors, Cell Surface/drug effects , Testicular Neoplasms/metabolism , Testis/drug effects , Animals , Estradiol/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Organ Size/drug effects , Rats , Receptors, Cell Surface/metabolism , Receptors, LH
9.
Cancer Res ; 44(3): 885-92, 1984 Mar.
Article in English | MEDLINE | ID: mdl-6318985

ABSTRACT

Possible influences of the tumor promoter 12-O-tetradecanoylphorbol-13-acetate (TPA) upon gonadal steroidogenesis were investigated in vitro. Granulosa cells from hypophysectomized, estrogen-treated rats were cultured for 2 days in medium containing 0.1 microM androstenedione. Follicle-stimulating hormone (FSH) treatment increased estrogen, progesterone, and 20 alpha-hydroxypregn-4-en-3-one production. Concomitant TPA treatment inhibited FSH-stimulated estrogen production by up to 95% [concentration that induced 50% inhibition of steroid production (ED50), 1.1 ng/ml]. TPA also inhibited FSH-stimulated progesterone (ED50, approximately 0.6 ng/ml) and 20 alpha-hydroxypregn-4-en-3-one (ED50, approximately 1.1 ng/ml) production. N6O2'-dibutyryl cyclic adenosine 3':5'-monophosphate increased steroidogenesis; however, cotreatment with TPA blocked progestin but not estrogen production. The TPA inhibition of progestin biosynthesis was accompanied by decreases in FSH-stimulated pregnenolone biosynthesis and 3 beta-hydroxysteroid dehydrogenase activity without decreasing the activity of 20 alpha-hydroxysteroid dehydrogenase. In primary cultures of rat testicular cells, human chorionic gonadotropin treatment increased testosterone production 44-fold, whereas concomitant treatment with TPA inhibited testosterone production by up to 86% (ED50, 10 ng/ml). Cholera toxin and N6O2'-dibutyryl cyclic adenosine 3':5'-monophosphate also increased testosterone production, while the actions of these agents were decreased by TPA. The TPA suppression of testosterone production was associated with a decrease in accumulation of 17 alpha-hydroxyprogesterone and androstenedione and an increase in progesterone production, suggesting a specific inhibition of 17 alpha-hydroxylase and 17,20-lyase activities. These results demonstrate the inhibitory effects of a tumor promoter upon gonadotropin-stimulated steroidogenesis by cultured rat granulosa and Leydig cells through specific regulation of steroidogenic enzymes. Additional studies may assist in further elucidation of cellular mechanisms associated with carcinogenesis and steroidogenesis.


Subject(s)
Gonadal Steroid Hormones/biosynthesis , Granulosa Cells/metabolism , Phorbols/toxicity , Steroids/biosynthesis , Testis/metabolism , Tetradecanoylphorbol Acetate/toxicity , Animals , Bucladesine/pharmacology , Estrogens/biosynthesis , Female , Follicle Stimulating Hormone/pharmacology , Granulosa Cells/drug effects , In Vitro Techniques , Kinetics , Male , Progesterone/biosynthesis , Rats , Rats, Inbred Strains , Testis/drug effects
10.
J Endocrinol ; 187(3): 333-8, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16423812

ABSTRACT

Recent advances in comparative genomics allow a new paradigm for hormonal research. At the centennial of the first use of the term hormone by Ernest Starling, we reflected on the changing approaches in elucidating hormonal signaling mechanisms and highlighted the inadequacy of the term endocrinology, implying remote activation, to describe the diverse modes of hormone actions. Several examples were presented to underscore the power of comparative genomics in the identification of new polypeptide hormones, receptors, and signaling pathways. We propose the use of the term hormonology to more accurately reflect the expanding boundaries of the discipline.


Subject(s)
Genomics , Hormones/physiology , Bone Morphogenetic Protein 15 , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/physiology , Cell Communication/genetics , Cell Communication/physiology , Genome, Human , Growth Differentiation Factor 9 , Hormones/blood , Hormones/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/physiology , Ligands , Peptide Hormones/genetics , Peptide Hormones/physiology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/physiology
11.
Trends Endocrinol Metab ; 3(5): 164-70, 1992 Jul.
Article in English | MEDLINE | ID: mdl-18407096

ABSTRACT

The anterior pituitary hormones, luteinizing hormone (LH) and follicle-stimulating hormone (FSH), act upon the ovary and testis via occupancy of specific cell membrane receptors, resulting in increased cAMP production, steroidogenesis, and expression of differentiation-related genes. Recent cloning of the cDNAs for LH and FSH receptors allows the analysis of mRNA levels for these receptors in gonadal tissues. This review summarizes progress in elucidating the molecular basis of LH and FSH receptor gene regulation in the ovary and testis during different physiologic states.

12.
Trends Endocrinol Metab ; 8(5): 207-13, 1997 Jul.
Article in English | MEDLINE | ID: mdl-18406807

ABSTRACT

Using the ovary as a model system for studying the hormonal regulation of apoptosis, recent studies have revealed that the survival of growing follicles is under the regulation of a complex array of hormones through endocrine, paracrine, autocrine, or juxtacrine mechanism in a development-dependent manner. More effort is needed, however, to identify tissue-specific factors required for the survival of ovarian somatic and germ cells at specific stage of development. New insights based on characterization of conserved apoptotic effectors, both extracellular and intracellular, have suggested that apoptosis in ovarian cells may be mediated by apoptotic programs common to other cells but using specific members of the death domain proteins as well as ced-9/Bcl-2 and ced-3/ICE caspase families of genes. Future studies may provide new therapeutic modalities for different ovarian diseases caused by aberrant regulation of apoptosis in ovarian cells, including premature ovarian failure and polycystic ovarian syndrome. (Trends Endocrinol Metab 1997;8:207-213). (c) 1997, Elsevier Science Inc.

13.
Mol Endocrinol ; 2(9): 854-61, 1988 Sep.
Article in English | MEDLINE | ID: mdl-3139993

ABSTRACT

FSH and GnRH both stimulate rat granulosa cells to produce tissue-type plasminogen activator (tPA). We have studied the molecular mechanisms involved in the action of these hormones by measuring tPA mRNA levels in primary cultures of rat granulosa cells. When granulosa cells were cultured in the presence of FSH or GnRH the level of tPA mRNA was increased 20- and 12-fold, respectively. The induction of tPA mRNA by FSH and GnRH was additive and the kinetics of induction differed. The effect of FSH could be mimicked by bromo-cAMP or forskolin, and was drastically enhanced by cotreatment with the phosphodiesterase inhibitor 1-methyl-3-isobutylxanthine. These findings are consistent with the notion that FSH mediates its effect through the protein kinase A pathway. GnRH is believed to augment phospholipid turnover in granulosa cells, leading to the activation of the protein kinase C pathway. Like GnRH, the protein kinase C activator phorbol myristate acetate also induced tPA mRNA in granulosa cells. In the presence of the protein synthesis inhibitor, cycloheximide, FSH-stimulated tPA message levels were enhanced by 30-fold, revealing superinduction of tPA mRNA levels by this pathway. In contrast the induction of tPA mRNA by GnRH was inhibited by cycloheximide indicating that the synthesis of an intermediate protein is required for the GnRH effect. Our data suggest that FSH and GnRH increase the tPA mRNA levels by two distinct pathways in cultured granulosa cells, providing a model system for studying the hormonal regulation of tPA gene expression.


Subject(s)
Follicle Stimulating Hormone/pharmacology , Gonadotropin-Releasing Hormone/pharmacology , Granulosa Cells/metabolism , RNA, Messenger/metabolism , Tissue Plasminogen Activator/genetics , Animals , Cycloheximide/pharmacology , DNA Probes , Dactinomycin/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation , Female , Granulosa Cells/drug effects , Protein Biosynthesis/drug effects , Protein Kinase C/metabolism , Protein Kinases/metabolism , Rats , Rats, Inbred Strains , Time Factors , Transcription, Genetic/drug effects
14.
Mol Endocrinol ; 12(9): 1432-40, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9731710

ABSTRACT

Using the yeast two-hybrid protein-protein interaction system to search for genes capable of forming dimers with the antiapoptotic protein Mcl-1, we have isolated BOD (Bcl-2-related ovarian death agonist) from an ovarian fusion cDNA library. The three variants of BOD (long, medium, and short) have an open reading frame of 196, 110, and 93 amino acids, respectively; all of them contain a consensus Bcl-2 homology 3 (BH3) domain but lack other BH domains found in channel-forming Bcl-2 family proteins. In the yeast cell assay, BOD interacts with diverse antiapoptotic Bcl-2 proteins [Mcl-1, Bcl-2, Bcl-xL, Bcl-w, Bfl-1, and Epstein-Barr virus (EBV) BHRF-1] but not with different proapoptotic Bcl-2 proteins (BAD, Bak, Bok, and Bax). After overexpression in mammalian Chinese hamster ovary (CHO) cells, BOD induces apoptosis that can be prevented by the baculoviral caspase inhibitor P35. The cell-killing activity of BOD is also antagonized in cells cotransfected with the antiapoptotic Bcl-w protein, which showed high affinity for BOD in the two-hybrid assay. Furthermore, mutagenesis studies showed that BOD mutants with alterations in the BH3 domain lose cell-killing ability, suggesting that the BH3 domain is important for the mediation of cell killing by BOD. BOD mRNA is ubiquitously expressed in ovary and multiple other tissues. The BOD gene is also conserved in diverse mammalian species. Identification of BOD expands the group of proapoptotic Bcl-2 proteins that only contains the BH3 domain and allows future elucidation of the intracellular mechanism for apoptosis regulation in ovary and other tissues.


Subject(s)
Apoptosis , Carrier Proteins/metabolism , Membrane Proteins , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins , Alternative Splicing , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins , Bcl-2-Like Protein 11 , CHO Cells , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cricetinae , Dimerization , Humans , Molecular Sequence Data , Myeloid Cell Leukemia Sequence 1 Protein , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/metabolism , Rats
15.
Mol Endocrinol ; 6(11): 1942-50, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1480180

ABSTRACT

Recent biochemical studies have suggested that apoptotic cell death is the molecular mechanism underlying the degeneration of ovarian follicles during atresia. Using a sensitive autoradiographic method for the detection of DNA fragmentation, we studied apoptosis in ovarian granulosa cells or intact follicles placed in serum-free culture as model systems to elucidate the hormonal regulation of atresia. Immature rats (25 days old) were primed for 2 days with 10 IU equine CG to induce a homogeneous population of mature preovulatory follicles. Granulosa cells isolated from these follicles contained predominantly intact high mol wt DNA. However, a time-dependent, spontaneous onset of internucleosomal DNA fragmentation characteristic of apoptotic cell death occurred in granulosa cells during culture. Treatment of granulosa cells with epidermal growth factor (EGF), transforming growth factor-alpha (TGF alpha), or basic fibroblast growth factor (bFGF) inhibited the spontaneous onset of apoptotic DNA cleavage found during culture by 40-60%. In contrast, insulin-like growth factor I, insulin, TGF beta and tumor necrosis factor-alpha were ineffective. Likewise, activation of the protein kinase A or C pathways with forskolin or phorbol 12-myristate 13-acetate, respectively, did not prevent the onset of DNA fragmentation, although inclusion of a tyrosine kinase inhibitor (genistein) completely blocked the ability of EGF, TGF alpha, and bFGF to suppress apoptosis in granulosa cells. Similar to cultured granulosa cells, a spontaneous onset of apoptosis was also observed to occur in isolated preovulatory follicles during culture. Furthermore, treatment of follicles with EGF or bFGF inhibited the spontaneous initiation of apoptosis, and the suppressive effects of these growth factors were also attenuated by co-treatment with genistein.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Apoptosis/drug effects , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/pharmacology , Granulosa Cells/drug effects , Ovarian Follicle/drug effects , Protein-Tyrosine Kinases/physiology , Animals , Cells, Cultured , Colforsin/pharmacology , Enzyme Activation , Female , Genistein , Isoflavones/pharmacology , Protein Kinase C/metabolism , Protein Kinases/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Transforming Growth Factor alpha/pharmacology
16.
Mol Endocrinol ; 5(3): 397-403, 1991 Mar.
Article in English | MEDLINE | ID: mdl-1890990

ABSTRACT

Down-regulation of plasma membrane receptors by homologous hormones has been found in diverse cell types. In testicular Leydig and ovarian luteal cells, treatment with LH/hCG decreases LH receptor content. Although suppression of LH-binding sites may result from ligand-induced receptor internalization, sequestration, and/or phosphorylation, the gonadotropins may also regulate receptor mRNA levels. We examined the regulation of testis LH receptor mRNAs in adult rats that received 10 or 200 IU hCG, using cRNA probes derived from the 5' extracellular domain (EC) or the 3' transmembrane domain (TM) of the rat receptor cDNA. Probe EC hybridized to predominant signals of 7 and 1.8 kilobases (kb) and weaker signals of 4.2 and 2.5 kb. However, probe TM hybridized to the three larger forms of the LH receptor mRNA, but not to the 1.8-kb species, suggesting that the latter form lacks the transmembrane domain. After 6 and 12 h of treatment with 200 or 10 IU hCG, respectively, hybridization to the larger mRNA species decreased by more than 60%, preceding decreases in testicular [125I]hCG binding. These transcripts were further inhibited (greater than 93%) between 24-72 h after hCG treatment and returned to 40% and 100% of control levels by days 6 and 9, respectively. In contrast, the truncated 1.8-kb LH receptor transcript was not affected by hCG treatment, indicating a differential suppressive effect of the ligand on its receptor mRNA levels. In the ovary, hybridization to probe EC revealed four transcripts with similar sizes as those found in the testes, with a predominant 7-kb species.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Chorionic Gonadotropin/pharmacology , Down-Regulation , Luteinizing Hormone/pharmacology , Ovary/metabolism , Receptors, LH/genetics , Testis/metabolism , Animals , Blotting, Northern , Cells, Cultured , DNA Probes , Female , Leydig Cells/metabolism , Ligands , Male , Ovary/drug effects , Pregnancy , RNA Probes , RNA, Messenger/biosynthesis , RNA, Messenger/metabolism , Rats , Testis/drug effects
17.
Mol Endocrinol ; 11(11): 1659-68, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9328348

ABSTRACT

Receptors for the glycoprotein hormones, LH/CG, FSH, and TSH, are a unique subclass of the seven-transmembrane, G protein-coupled proteins with a large N-terminal extracellular (ecto-) domain. Although ecto-domains of gonadotropin receptors confer ligand binding, expression of soluble binding proteins has been difficult. We fused the ecto-domains of LH or FSH receptors to the single-transmembrane domain of CD8 and found that hybrid proteins anchored on the cell surface retained high-affinity ligand binding. Inclusion of a junctional thrombin cleavage site in the hybrids allowed generation of soluble receptor fragments that interfered with gonadotropin binding to their receptors and blocked cAMP production stimulated by gonadotropins. Cross-linking analyses confirmed the formation of high molecular weight complexes between receptor ecto-domains and their specific ligands. A similar approach also generated a soluble TSH receptor fragment capable of blocking TSH-induced signal transduction. When administered to rats, the soluble FSH receptor fragment retarded testis growth and induced testis cell apoptosis. These findings demonstrate the feasibility of generating ligand-binding regions of glycoprotein hormone receptors to selectively neutralize actions of gonadotropins and TSH, thus allowing future design of novel contraceptives and management of different gonadal and thyroid dysfunction. The present study represents the first successful derivation of soluble, ligand-binding domains from glycoprotein hormone receptors as functional antagonists. Similar approaches could allow generation of ecto-domains of related receptors to neutralize actions of ligands or receptor antibodies and to facilitate structural-functional analysis.


Subject(s)
Peptide Fragments/pharmacology , Receptors, FSH/agonists , Receptors, LH/agonists , Receptors, Thyrotropin/agonists , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , CD8 Antigens/biosynthesis , CD8 Antigens/chemistry , CD8 Antigens/genetics , Cell Line , Contraceptive Agents/chemistry , Contraceptive Agents/pharmacology , Cyclic AMP/biosynthesis , Drug Design , GTP-Binding Proteins/physiology , Genetic Vectors , Humans , Ligands , Male , Mutagenesis, Site-Directed , Nucleopolyhedroviruses/genetics , Peptide Fragments/chemistry , Protein Binding , Protein Conformation , Rats , Rats, Sprague-Dawley , Receptors, FSH/chemistry , Receptors, FSH/genetics , Receptors, LH/chemistry , Receptors, LH/genetics , Receptors, Thyrotropin/chemistry , Receptors, Thyrotropin/genetics , Recombinant Fusion Proteins/drug effects , Spodoptera , Structure-Activity Relationship , Testis/drug effects , Testis/pathology
18.
Mol Endocrinol ; 15(5): 681-94, 2001 May.
Article in English | MEDLINE | ID: mdl-11328851

ABSTRACT

The cystine knot three-dimensional structure is found in many extracellular molecules and is conserved among divergent species. The identification of proteins with a cystine knot structure is difficult by commonly used pairwise alignments because the sequence homology among these proteins is low. Taking advantage of complete genome sequences in diverse organisms, we used a complementary approach of pattern searches and pairwise alignments to screen the predicted protein sequences of five model species (human, fly, worm, slime mold, and yeast) and retrieved proteins with low sequence homology but containing a typical cystine knot signature. Sequence comparison between proteins known to have a cystine knot three-dimensional structure (transforming growth factor-beta, glycoprotein hormone, and platelet-derived growth factor subfamily members) identified new crucial amino acid residues (two hydrophilic amino acid residues flanking cysteine 5 of the cystine knot). In addition to the well known members of the cystine knot superfamily, novel subfamilies of proteins (mucins, norrie disease protein, von Willebrand factor, bone morphogenetic protein antagonists, and slit-like proteins) were identified as putative cystine knot-containing proteins. Phylogenetic analysis revealed the ancient evolution of these proteins and the relationship between hormones [e.g. transforming growth factor-beta (TGFbeta)] and extracellular matrix proteins (e.g. mucins). They are absent in the unicellular yeast genome but present in nematode, fly, and higher species, indicating that the cystine knot structure evolved in extracellular signaling molecules of multicellular organisms. All data retrieved by this study can be viewed at http://hormone.stanford.edu/.


Subject(s)
Cystine/chemistry , Hormones/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Protein Conformation , Protein Structure, Tertiary , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction
19.
Mol Endocrinol ; 12(12): 1830-45, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9849958

ABSTRACT

The receptors for LH, FSH, and TSH belong to the large G protein-coupled, seven-transmembrane (TM) protein family and are unique in having a large N-terminal extracellular (ecto-) domain containing leucine-rich repeats important for interaction with the glycoprotein ligands. We have identified two new leucine-rich repeat-containing, G protein-coupled receptors and named them as LGR4 and LGR5, respectively. The ectodomains of both receptors contain 17 leucine-rich repeats together with N- and C-terminal flanking cysteine-rich sequences, compared with 9 repeats found in known glycoprotein hormone receptors. The leucine-rich repeats in LGR4 and LGR5 are arrays of 24 amino acids showing similarity to repeats found in the acid labile subunit of the insulin-like growth factor (IGF)/IGF binding protein complexes as well as slit, decorin, and Toll proteins. The TM region and the junction between ectodomain and TM 1 are highly conserved in LGR4, LGR5, and seven other LGRs from sea anemone, fly, nematode, mollusk, and mammal, suggesting their common evolutionary origin. In contrast to the restricted tissue expression of gonadotropin and TSH receptors in gonads and thyroid, respectively, LGR4 is expressed in diverse tissues including ovary, testis, adrenal, placenta, thymus, spinal cord, and thyroid, whereas LGR5 is found in muscle, placenta, spinal cord, and brain. Hybridization analysis of genomic DNA indicated that LGR4 and LGR5 genes are conserved in mammals. Comparison of overall amino acid sequences indicated that LGR4 and LGR5 are closely related to each other but diverge, during evolution, from the homologous receptor found in snail and the mammalian glycoprotein hormone receptors. The identification and characterization of new members of the LGR subfamily of receptor genes not only allow future isolation of their ligands and understanding of their physiological roles but also reveal the evolutionary relationship of G protein-coupled receptors with leucine-rich repeats.


Subject(s)
GTP-Binding Proteins/metabolism , Leucine , Receptors, Cell Surface/chemistry , Receptors, G-Protein-Coupled , Repetitive Sequences, Amino Acid , Amino Acid Sequence , Animals , Conserved Sequence , DNA/isolation & purification , Female , Humans , Male , Molecular Sequence Data , Organ Specificity , Phylogeny , Receptors, Cell Surface/genetics , Receptors, FSH/genetics , Receptors, LH/chemistry , Receptors, Thyrotropin/chemistry , Sequence Homology
20.
Mol Endocrinol ; 11(12): 1858-67, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9369453

ABSTRACT

Apoptosis and survival of diverse cell types are under hormonal control, but intracellular mechanisms regulating cell death are unclear. The Bcl-2/Ced-9 family of proteins contains conserved Bcl-2 homology regions that mediate the formation of homo- or heterodimers important for enhancing or suppressing apoptosis. Unlike most other members of the Bcl-2 family, BAD (Bcl-xL/Bcl-2 associated death promoter), a death enhancer, has no C-terminal transmembrane domain for targeting to the outer mitochondrial membrane and nuclear envelope. We hypothesized that BAD, in addition to binding Bcl-xL and Bcl-2, may interact with proteins outside the Bcl-2 family. Using the yeast two-hybrid system to search for BAD-binding proteins in an ovarian fusion cDNA library, we identified multiple cDNA clones encoding different isoforms of 14-3-3, a group of evolutionally conserved proteins essential for signal transduction and cell cycle progression. Point mutation of BAD in one (S137A), but not the other (S113A), putative binding site found in diverse 14-3-3 interacting proteins abolished the interaction between BAD and 14-3-3 without affecting interactions between BAD and Bcl-2. Because the S137A BAD mutant presumably resembles an underphosphorylated form of BAD, we used this mutant to screen for additional BAD-interacting proteins in the yeast two-hybrid system. P11, a nerve growth factor-induced neurite extension factor and member of the calcium-binding S-100 protein family, interacted strongly with the mutant BAD but less effectively with the wild type protein. In Chinese hamster ovary (CHO) cells, transient expression of wild type BAD or its mutants increased apoptotic cell death, which was blocked by cotransfection with the baculovirus-derived cysteine protease inhibitor, P35. Cotransfection with 14-3-3 suppressed apoptosis induced by wild type or the S113A mutant BAD but not by the S137A mutant incapable of binding 14-3-3. Furthermore, cotransfection with P11 attenuated the proapoptotic effect of both wild type BAD and the S137A mutant. For both 14-3-3 and P11, direct binding to BAD was also demonstrated in vitro. These results suggest that both 14-3-3 and P11 may function as BAD-binding proteins to dampen its apoptotic activity. Because the 14-3-3 family of proteins could interact with key signaling proteins including Raf-1 kinase, protein kinase C, and phosphatidyl inositol 3 kinase, whereas P11 is an early response gene induced by the neuronal survival factor, nerve growth factor, the present findings suggest that BAD plays an important role in mediating communication between different signal transduction pathways regulated by hormonal signals and the apoptotic mechanism controlled by Bcl-2 family members.


Subject(s)
Apoptosis/drug effects , Carrier Proteins/drug effects , Nerve Growth Factors/pharmacology , Nerve Growth Factors/physiology , Proteins/pharmacology , Tyrosine 3-Monooxygenase , 14-3-3 Proteins , Animals , Apoptosis/genetics , Binding Sites/drug effects , Binding Sites/genetics , CHO Cells , Carrier Proteins/biosynthesis , Carrier Proteins/physiology , Cricetinae , Female , Mutagenesis, Site-Directed , Nerve Growth Factors/metabolism , Nucleopolyhedroviruses , Protein Binding/drug effects , Proteins/metabolism , Rats , Rats, Sprague-Dawley , Viral Proteins/pharmacology , Yeasts/genetics , bcl-Associated Death Protein
SELECTION OF CITATIONS
SEARCH DETAIL