Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Bone ; 78: 62-70, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25937185

ABSTRACT

Exercise is vital for maintaining bone strength and architecture. Follistatin-like 3 (FSTL3), a member of follistatin family, is a mechanosensitive protein upregulated in response to exercise and is involved in regulating musculoskeletal health. Here, we investigated the potential role of FSTL3 in exercise-driven bone remodeling. Exercise-dependent regulation of bone structure and functions was compared in mice with global Fstl3 gene deletion (Fstl3-/-) and their age-matched Fstl3+/+ littermates. Mice were exercised by low-intensity treadmill walking. The mechanical properties and mineralization were determined by µCT, three-point bending test and sequential incorporation of calcein and alizarin complexone. ELISA, Western-blot analysis and qRT-PCR were used to analyze the regulation of FSTL3 and associated molecules in the serum specimens and tissues. Daily exercise significantly increased circulating FSTL3 levels in mice, rats and humans. Compared to age-matched littermates, Fstl3-/- mice exhibited significantly lower fracture tolerance, having greater stiffness, but lower strain at fracture and yield energy. Furthermore, increased levels of circulating FSTL3 in young mice paralleled greater strain at fracture compared to the lower levels of FSTL3 in older mice. More significantly, Fstl3-/- mice exhibited loss of mechanosensitivity and irresponsiveness to exercise-dependent bone formation as compared to their Fstl3+/+ littermates. In addition, FSTL3 gene deletion resulted in loss of exercise-dependent sclerostin regulation in osteocytes and osteoblasts, as compared to Fstl3+/+ osteocytes and osteoblasts, in vivo and in vitro. The data identify FSTL3 as a critical mediator of exercise-dependent bone formation and strengthening and point to its potential role in bone health and in musculoskeletal diseases.


Subject(s)
Bone and Bones/metabolism , Follistatin-Related Proteins/physiology , Gene Expression Regulation , Osteoblasts/cytology , Osteocytes/cytology , Adult , Aged , Animals , Anthraquinones/chemistry , Bone Remodeling , Enzyme-Linked Immunosorbent Assay , Exercise Test , Female , Fluoresceins/chemistry , Gene Deletion , Humans , Male , Mice , Mice, Inbred C57BL , Physical Conditioning, Animal , Rats , Rats, Sprague-Dawley , Stress, Mechanical , Time Factors , Up-Regulation , Walking , X-Ray Microtomography , Young Adult
2.
Endocrinology ; 142(8): 3426-34, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11459787

ABSTRACT

Follistatin-related protein is a recently discovered glycoprotein that is highly homologous in both primary sequence and exon/intron domain structure to the activin-binding protein, follistatin. We explored their potential for functional redundancy by investigating the relative affinities and kinetics of their interactions with activin, bone morphogenic protein-6, and bone morphogenic protein-7 and by exploring their expression and distribution in human tissues and cells. Follistatin and follistatin-related protein mRNA were ubiquitous by Northern analyses, although their sites of peak distribution differed, with follistatin-related protein and follistatin predominating in the placenta and ovary, respectively. Follistatin-related protein, like follistatin, preferentially bound activin with high affinity and in an essentially irreversible fashion. Although follistatin-related protein, like follistatin, possesses a signal sequence and no known nuclear localization signals, its secretion was undetectable in most cell lines by RIA. Intriguingly, follistatin-related protein was identified as a nuclear protein in human granulosa cells and all human cell lines tested. Furthermore, Western analyses of CHO cells transfected with human follistatin-related protein revealed this protein to reside within the insoluble nuclear protein fraction. We conclude that despite its remarkably high level of similarity to follistatin with regard to structure and activin binding kinetics, follistatin-related protein is a nuclear as well as a secretory protein that may perform distinct intracellular actions.


Subject(s)
Cell Nucleus/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Activins , Amino Acid Sequence/genetics , Animals , Blotting, Western , CHO Cells , Cricetinae , Follistatin , Follistatin-Related Proteins , Humans , Immunohistochemistry , Inhibins/metabolism , Kinetics , Molecular Sequence Data , RNA, Messenger/metabolism , Radioimmunoassay , Tissue Distribution
3.
J Clin Endocrinol Metab ; 86(1): 330-6, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11232020

ABSTRACT

Serum inhibin B rises across the luteal-follicular transition, whereas inhibin A does not increase until the late follicular phase of the menstrual cycle. To test the hypothesis that inhibin B is secreted from preantral and small antral follicles and that FSH and local growth factors differentially regulate inhibin B and inhibin A from these developing follicles, human ovaries were obtained after oophorectomy. Basal secretion of inhibin B and inhibin A was examined in intact preantral follicles in culture (n = 6). Basal secretion and regulation of inhibin B and inhibin A secretion by gonadotropins, androstenedione, activin A, insulin, and IGF-I were examined in cultured granulosa cells from small antral follicles (n = 21). Inhibin B secretion from preantral follicle cultures was detectable at baseline (range, 17-96 pg/mL), whereas inhibin A was not detectable. In contrast, both inhibin B and inhibin A were detectable in granulosa cell cultures from small antral follicles. In granulosa cells from small antral follicles, FSH (30 ng/mL) stimulated inhibin A 3-fold (10.5 +/- 2.2 to 32.5 +/- 8.3 IU/mL; P < 0.001), but not inhibin B secretion (1730 +/- 354 to 2314 +/- 532 pg/mL; P = NS). Likewise, cAMP (1 mmol/L) stimulated inhibin A 4-fold (16.6 +/- 4.3 to 62.5 +/- 21.9 IU/mL; P < 0.002), but not inhibin B secretion (2327 +/- 546 to 1877 +/- 377 pg/mL; P = NS). hCG (30 ng/mL) did not stimulate inhibin A or inhibin B. Androstenedione (10(-)(7) mol/L), activin (30 ng/mL), insulin (30 ng/mL), and insulin-like growth factor I (IGF-I; 100 ng/mL) alone did not stimulate inhibin A or inhibin B secretion. Further, FSH-stimulated inhibin A secretion was not augmented by androstenedione, activin, insulin, or IGF-I. In contrast, the combination of IGF-I and FSH was the only treatment that stimulated inhibin B secretion (1742 +/- 380 to 2881 +/- 731 pg/mL; P < 0.03). However, FSH in combination with IGF-I resulted in greater stimulation of inhibin A (340%) than inhibin B (65%). These findings demonstrate that inhibin B is secreted from developing preantral and small antral follicles, but is not directly stimulated by FSH. However, the combination of FSH and IGF-I enhanced inhibin B secretion. In contrast, inhibin A is not secreted from preantral follicles, but in small antral follicles FSH and cAMP stimulate inhibin A secretion. Further, FSH in combination with IGF-I results in a greater degree of stimulation of inhibin A than of inhibin B. These findings suggest that FSH and IGF-I differentially regulate inhibin A and inhibin B secretion. However, additional growth factors or increasing granulosa cell number may contribute to the preferential serum inhibin B increase across the luteal-follicular transition in the menstrual cycle.


Subject(s)
Follicle Stimulating Hormone/physiology , Granulosa Cells/metabolism , Growth Substances/physiology , Inhibins/metabolism , Prostatic Secretory Proteins , Adult , Female , Humans , In Vitro Techniques , Organ Size , Ovarian Follicle/anatomy & histology , Polycystic Ovary Syndrome/metabolism , Reference Values , Stimulation, Chemical
4.
J Biol Chem ; 276(21): 17718-26, 2001 May 25.
Article in English | MEDLINE | ID: mdl-11279126

ABSTRACT

Follistatin is recognized to be an important regulator of cellular differentiation and secretion through its potent ability to bind and bioneutralize activin with which it is colocalized in many tissue systems. The 288-residue follistatin molecule is comprised of a 63-residue N-terminal segment followed by three repeating 10-cysteine "follistatin domains" also represented in several extracellular matrix proteins. We have used chemical modifications and mutational analyses to define structural requirements for follistatin bioactivity that previously have not been investigated systematically. Mutant follistatins were stably expressed from Chinese hamster ovary cell cultures and assayed for activin binding in a solid-phase competition assay. Biological activities were determined by inhibition of activin-mediated transcriptional activity and by suppression of follicle-stimulating hormone secretion by cultured anterior pituitary cells. Deletion of the entire N-terminal domain, disruption of N-terminal disulfides, and deletion of the first two residues each reduced activin binding to <5 % of expressed wild-type follistatin and abolished the ability of the respective mutants to suppress activin-mediated responses in both bioassay systems. Hence, the three follistatin domains inherently lack the ability to bind or neutralize activin. Activin binding was impaired after oxidation of at least one tryptophan, at position 4, in FS-288. Mutation of Trp to Ala or Asp at either positions 4 or 36 eliminated activin binding and bioactivity. Mutation of a third hydrophobic residue, Phe-52, reduced binding to 20%, whereas substitutions for the individual Lys and Arg residues in the N-terminal region were tolerated. These results establish that hydrophobic residues within the N-terminal domain constitute essential activin-binding determinants in the follistatin molecule. The correlation among the effects of mutation on activin binding, activin transcriptional responses, and follicle-stimulating hormone secretion substantiates the concept that, at least in the pituitary, the biological activity of follistatin is attributable to its ability to bind and bioneutralize activin.


Subject(s)
Glycoproteins/metabolism , Inhibins/metabolism , Activins , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Cricetinae , Follistatin , Glycoproteins/genetics , Humans , Inhibins/genetics , Molecular Sequence Data , Mutation , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship
5.
J Clin Endocrinol Metab ; 85(9): 3319-30, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10999828

ABSTRACT

Previous studies of normal human ovaries suggest that inhibins, activins, and follistatin (FS) are produced in a stage-specific pattern indicative of intraovarian, autocrine/paracrine roles in regulating follicle development. However, these studies relied largely on surgical specimens and thus include little information about the menstrual cycle stage or dominant follicle status at the time follicles or ovaries were obtained. The purpose of this study was to 1) determine the pattern of intrafollicular hormone biosynthesis across antral follicle development in normal women, 2) compare hormone concentrations in dominant and nondominant follicles from the same ovary, and 3) examine the relationship between dominant follicle hormone content and circulating hormone levels. Intrafollicular estradiol, progesterone, and inhibin A concentrations increased significantly with follicle size or maturity, whereas significant inverse relationships were observed for androstenedione and the androstenedione/estradiol (A:E) ratio. In contrast, neither inhibin B, activin A, nor free FS varied consistently with size or maturity. Estradiol, progesterone, and inhibin A levels and A:E ratio were significantly lower in nondominant follicles compared to the dominant follicle aspirated from the same ovary. Although intrafollicular and serum concentrations of each hormone followed the same general pattern as follicles develop, the human follicular fluid/serum gradients changed during the follicular phase and were different for estradiol and inhibin A, suggesting the presence of stage-specific differences in pharmacodynamics. These results are consistent with the hypothesis that the orderly transition from an activin-dominant to an inhibin A/FS-dominant microenvironment is critical for dominant follicle development.


Subject(s)
Glycoproteins/metabolism , Growth Substances/metabolism , Inhibins/metabolism , Ovarian Follicle/physiology , Activins , Adult , Androstenedione/blood , Estradiol/blood , Female , Follistatin , Glycoproteins/blood , Growth Substances/blood , Humans , Inhibins/blood , Luteinizing Hormone/blood , Ovarian Follicle/anatomy & histology , Ovulation/physiology , Progesterone/blood
6.
Endocrinology ; 141(9): 3183-93, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10965889

ABSTRACT

A primary physiological function of follistatin is the binding and neutralization of activin, a transforming growth factor-beta family growth factor, and loss of function mutations are lethal. Despite the critical biological importance of follistatin's neutralization of activin, the structural basis of activin's binding to follistatin is poorly understood. The purposes of these studies were 1) to identify the primary sequence(s) within the N-terminal domain of the follistatin coding for activin binding, and 2) to determine whether activin binding to the native protein causes changes in other structural domains of follistatin. Synthetic peptide mimotopes identified within a 63-residue N-terminal domain two discontinuous sequences capable of binding labeled activin A. The first is located in a region (amino acids 3-26) of follistatin, a site previously identified by directed mutagenesis as important for activin binding. The second epitope, predicted to be located between amino acids 46 and 59, is newly identified. Although the sequences 3-26 and 46-59 code for activin binding, native follistatin only binds activin if disulfide bonding is intact. Furthermore, pyridylethylation of Cys residues followed by N-terminal sequencing and amino acid analysis revealed that all of the Cys residues in follistatin are involved in disulfide bonds and lack reactive free sulfhydryl groups. Specific ligands were used to probe the structural effects of activin binding on the other domains of the full-length molecule, comprised largely of the three 10-Cys follistatin module domains. No effects on ligand binding to follistatin-like module I or II were observed after the binding of activin A to native protein. In contrast, activin binding diminished recognition of domain III and enhanced that of the C domain by their respective monoclonal antibody probes, indicating an alteration of the antigenic structures of these regions. Thus, subsequent to activin binding, interactions are likely to occur between regions of follistatin located in different domains and separated by considerable lengths of linear sequence. Such interactions could have important functional significance with respect to the structural heterogeneity of naturally occurring follistatins.


Subject(s)
Glycoproteins/genetics , Inhibins/metabolism , Activins , Amino Acid Sequence , Antibodies, Monoclonal , Electrophoresis, Polyacrylamide Gel , Epitopes/genetics , Follistatin , Glycoproteins/biosynthesis , Heparitin Sulfate , Humans , Ligands , Molecular Sequence Data , Molecular Weight , Peptide Mapping , Protein Binding , Protein Conformation , Proteins/chemistry , Proteins/genetics , Sulfhydryl Compounds/chemistry
7.
Endocrinology ; 140(6): 2463-70, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10342830

ABSTRACT

The activin/follistatin system is implicated in growth and differentiation of various cell types. Follistatin (FS), through binding and neutralizing activin, plays a major role in the regulation of activin bioavailability. We previously reported that ovarian PA1 cells constitutively secrete FS and show a decreased proliferation rate in response to exogenous activin only if cell surface associated FS is first removed by heparin treatment. These observations suggest that cell-associated FS prevents exogenous activin from accessing its receptor. We hypothesized that cell surface FS would differentially regulate the bioavailability of endogenous and exogenous activin in these cells. To examine the effect of endogenous activin, PA1 cells were stably transfected with an activin betaA-subunit complementary DNA (cDNA). The proliferation rate of five activin-secreting clones was measured by [3H]thymidine incorporation and compared with the proliferation rate of untransfected cells. In clones secreting levels of activin ranging from 22.6 +/- 7.1 to 42.4 +/- 9.9 ng/ml, proliferation was decreased by 31-72% at 96 h of culture, whereas one cell line secreting lower levels of activin (0.4 +/- 0.1 ng/ml) proliferated similarly to the untransfected cells, in which activin was not detectable. To further assess activin signaling, wild-type PA1 cells and activin-secreting clones were transiently transfected with an activin response element-luciferase reporter construct. Basal luciferase activity was 6-fold higher in activin-secreting clones than in wild-type PA1 cells. Exogenous activin (100 ng/ml) increased the transcriptional response of wild-type PA1 cells by 3-fold but did not increase reporter activity in activin secreting clones. Interestingly, the transcriptional response in activin secreting clones was always greater than the basal or activin-stimulated response in wild-type cells. Furthermore, we found that FS was removed from the cell surface by lipofectamine used for these transfections. Therefore, these results show that activation of the luciferase reporter gene occurs under conditions in which proliferation is affected, suggesting that the antiproliferative effect of activin could be due to a direct stimulation of activin signaling pathways. In summary, as opposed to exogenous activin, endogenous activin decreased proliferation of PA1 cells even in the presence of cell surface associated FS. These results are consistent with a model in which FS acts as a barrier for exogenous (endocrine-paracrine) but not for endogenous (autocrine) activin. In addition, the higher PA1 cell responsiveness to endogenous compared with exogenous activin, suggests that activin overexpression in PA1 cells may up-regulate an activin signaling component, or down-regulate an activin signaling inhibitor.


Subject(s)
Glycoproteins/physiology , Inhibins/physiology , Ovarian Neoplasms/pathology , Teratocarcinoma/pathology , Activins , Cell Division , Female , Follistatin , Humans , Inhibins/pharmacology , Transcriptional Activation , Transfection , Tumor Cells, Cultured
8.
Biol Reprod ; 59(4): 807-12, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9746729

ABSTRACT

Inhibin, activin, and follistatin (FS) are gonadal proteins that appear to have a role in regulating folliculogenesis through possible paracrine and/or autocrine interactions. To further examine the potential role of activin in oocyte-granulosa cell communication, we developed a sensitive reverse transcription-polymerase chain reaction protocol to analyze mRNA for the alpha, betaA, and betaB inhibin/activin subunits, FS, and the four activin receptor subtypes in individual human and mouse oocytes. The resulting expression pattern was further compared to that in human cumulus granulosa cells. Our results indicate that neither ssA nor betaB mRNA was detectable in any human or mouse oocyte, that alpha subunit was marginally present in some of the human oocytes, and that FS mRNA was detectable in human but not mouse oocytes. On the other hand, inhibin/activin subunit and FS mRNAs were abundantly expressed in cumulus cells. In addition, mRNAs for all four activin receptor subtypes (ActRIA, ActRIB, ActRIIA, and ActRIIB) were easily detectable in both oocytes and granulosa cells and appeared to be differentially expressed in oocytes during nuclear maturation. Finally, RNAs for both zona pellucida 3 and growth-differentiation factor-9, which were originally used as oocyte-specific markers, were detected in human but not mouse cumulus cells, although at lower levels than observed in oocytes. Taken together with previous studies, these results indicate that oocytes may be capable of responding to, but not synthesizing, activin.


Subject(s)
Glycoproteins/biosynthesis , Granulosa Cells/physiology , Growth Substances/biosynthesis , Inhibins/biosynthesis , Oocytes/physiology , Paracrine Communication/physiology , Protein Serine-Threonine Kinases/biosynthesis , RNA, Messenger/biosynthesis , Receptors, Growth Factor/biosynthesis , Activin Receptors , Activins , Animals , Female , Follistatin , Glycoproteins/genetics , Growth Substances/genetics , Humans , In Vitro Techniques , Inhibins/genetics , Mice , Oocytes/metabolism , Paracrine Communication/genetics , Pregnancy , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , Receptors, Growth Factor/genetics
9.
Gynecol Oncol ; 69(1): 23-31, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9570994

ABSTRACT

OBJECTIVES: Inhibins and activins are related gonadal peptides with opposing biologic actions on gonadotropin regulation, cell differentiation, and proliferation. The previous study of activin in ovarian cancer cell lines suggests that activin may promote growth of ovarian cancer. Elevated serum inhibin levels were also found in ovarian cancer patients; however, the source of elevated inhibin is unknown. This study is designed to examine the expression of inhibin and activin subunits as well as activin receptor in primary ovarian epithelial tumors to explore their role in the process of ovarian epithelial tumorigenesis. METHODS: The protein and mRNA expression of alpha and betaA subunits of inhibin/activin as well as of activin receptor mRNA were examined with immunohistochemistry (IHC) and reverse transcription-polymerase chain reaction (RT-PCR) in 112 ovarian carcinomas. Cases included 59 serous, 23 endometrioid, 16 mucinous, 9 clear cell, and 5 undifferentiated carcinomas. We also tested normal ovary and benign and borderline ovarian tumors for comparison. These included 17 ovarian surface epithelial samples, 6 serous and 5 mucinous cystadenomas, and 9 serous and 7 mucinous tumors of low malignant potential. A total of 139 ovarian tumors were analyzed by IHC and a total of 63 ovarian tumor samples were tested by RT-PCR. RESULTS: Inhibin alpha subunit expression was found in 47% of ovarian surface epithelia and focal alpha immunoreactivity was seen in tumor stroma, but was not found in the epithelial component of ovarian cystadenomas, tumors of low malignant potential (LMP), or carcinomas. Activin betaA subunit was expressed in 93% of surface epithelia, in the epithelial component of all cystadenomas, in 81% of LMP tumors, and in 72% of carcinomas, but not in tumor stroma. Activin expression did not correlate with histologic grades, tumor types, and surgical stages. Activin receptor type I and II mRNA-amplified products were found in virtually all the surface epithelial samples and ovarian tumors. CONCLUSIONS: The data suggest that imbalanced expression of inhibin and activin subunits in ovarian surface epithelium may represent an early event which leads to epithelial proliferation. Unopposed betaA and activin receptor expression in epithelial compartment of ovarian tumors suggest that activin may be available as autocrine and/or paracrine factors in ovarian epithelial tumors. But exact roles of inhibin and activin in ovarian epithelial tumors remain to be defined.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma/chemistry , Inhibins/biosynthesis , Ovarian Neoplasms/chemistry , Receptors, Growth Factor/analysis , Activin Receptors , Activins , Adult , Carcinoma/genetics , Carcinoma/metabolism , Epithelium/metabolism , Female , Gene Expression , Humans , Immunohistochemistry , Inhibins/analysis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Polymerase Chain Reaction , RNA, Messenger/biosynthesis
10.
Endocrinology ; 139(3): 1147-55, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9492049

ABSTRACT

Activin, a member of the transforming growth factor-beta superfamily of growth and differentiation factors, has a number of actions in embryonic as well as adult tissues. These actions are mediated via a family of receptors containing two subtypes and at least two members of each subtype. Recent evidence demonstrates that activin-responsive cell lines containing different subsets of these receptors are valuable models for dissecting functional relationships among receptor subtype, signal transduced, and response obtained. TT cells, derived from a p53(-/-)/alpha-inhibin(-/-) mouse testicular tumor, respond to activin by proliferating, a response that can be inhibited by follistatin (FS) treatment. Using semiquantitative RT-PCR methods, we characterized steady state messenger RNA (mRNA) levels for the inhibin/activin subunits, FS, and activin receptor subtypes under basal conditions and in the presence of activin or FS. These cells produced ample immunoreactive activin A and FS, necessitating higher treatment doses to observe any modulation of cellular proliferation. Furthermore, in the presence of exogenous activin, mRNA levels for activin receptor type IIA (ACTRIIA) and betaA were significantly and profoundly suppressed. In addition, both ACTR1B and ACTRIIB were detectable and down-regulated by exogenous activin, although not to the degree observed for ACTRIIA and betaA. Finally, activin treatment at the higher doses, which decreased activin receptor mRNA levels, resulted in inhibition of cellular proliferation. Taken together with previous observations, our results support the model that these tumor cells respond to an autocrine activin signal by proliferating, whereas exogenous or excess activin results in down-regulation of activin receptor and activin biosynthesis, suggesting a potential autocrine/paracrine mechanism by which activin can modulate its own signal.


Subject(s)
Inhibins/physiology , RNA, Messenger/analysis , Receptors, Growth Factor/genetics , Testicular Neoplasms/metabolism , Activin Receptors , Activins , Animals , Cell Division , Follistatin , Glycoproteins/genetics , Glycoproteins/metabolism , Male , Mice , Testicular Neoplasms/pathology , Tumor Cells, Cultured
11.
J Clin Endocrinol Metab ; 83(3): 851-8, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9506739

ABSTRACT

Follistatin (FS) is a monomeric protein that binds and regulates the bioavailability of activin. Previously, we found circulating levels of total FS to be similar in men and cycling women. Because relative amounts of activin-bound and free FS are important considerations in determining activin bioavailability, we asked here whether the relative proportions of these two changed during different physiologic states. For this, we developed a two-site, solid-phase, immunochemiluminescent assay for free FS. The assay recognizes the 288 or 315 amino acid variants of human FS and has a detectable limit of 1 ng/mL. Inhibin, transforming growth factor-beta, or alpha-2-macroglobulin do not cross-react or interfere in this assay. Preincubation of FS with activin results in dose-dependent loss of immunoreactivity, confirming specificity of the assay for free FS. Human follicular fluid, pituitary extract, and serum with added FS dilute parallel with the recombinant human FS-288 standard. Recovery of recombinant human FS-288 from serum is quantitative. Using this assay, we found circulating concentrations of free FS to be at or below the detection limit of the assay throughout the menstrual cycle. Comparison of circulating total and free FS levels in postmenopausal or cycling women and normal men suggested that at least 90% is activin-bound. In contrast, measurable quantities of free FS were found in follicular fluid and pituitary extracts. The results of this study, showing that most circulating FS is normally activin-bound, argue against an endocrine role for FS and suggest that a major role of circulating FS is to bind and neutralize the bioactivity of circulating activin. The roles of FS as a local autocrine or paracrine regulator of activin in target tissues, where FS exists in free form, or as an endocrine regulator in human pathophysiology, warrants further investigation.


Subject(s)
Glycoproteins/metabolism , Inhibins/metabolism , Menstrual Cycle/blood , Activins , Female , Follistatin , Glycoproteins/blood , Glycoproteins/pharmacology , Humans , Immunoassay , Inhibins/blood , Luminescent Measurements , Male , Recombinant Proteins , Reference Values , Sensitivity and Specificity
12.
J Clin Endocrinol Metab ; 82(11): 3720-7, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9360531

ABSTRACT

Activin induces proliferation in epithelial ovarian carcinoma cell lines, whereas follistatin (FS), an activin binding protein, inhibits this action. To test the hypothesis that activin production, in excess of inhibin and FS, results in cell proliferation in epithelial ovarian tumors, messenger RNA (mRNA) expression of the activin family of proteins, FS, and activin type I and II receptors was examined in 25 primary epithelial ovarian tumors and tumor epithelium in culture (n = 7) using RT-PCR. Activin A was measured in the serum of ovarian cancer patients, and activin A, total inhibin, and FS protein secretion was measured from primary epithelial tumors in vitro. The effect of activin and FS on cell proliferation was assessed by measuring [3H]thymidine incorporation. All results were compared with normal ovarian epithelium. All epithelial ovarian tumors expressed mRNA for the alpha, beta A, and beta B subunits; FS 288 and 315; and the activin type IA, IB, II, and IIB receptors. beta A mRNA expression, as assessed using semiquantitative RT-PCR, was 3-fold greater in cultured tumor epithelium than in primary tumors (band density 0.86 +/- 0.17 vs. 0.28 +/- 0.09; P < 0.01). In addition, beta A mRNA was abundantly expressed in normal epithelium in culture (n = 2), whereas only trace amounts were seen in 2/9 primary epithelial samples. Activin protein was secreted by 24/25 primary epithelial ovarian tumors (range 0.2-155.8 ng/mL). In contrast, total inhibin was secreted by only 2/25 (range 0.01-0.92 ng/mL), whereas free FS was not detectable in the medium of any tumor (< 0.5 ng/mL). Treatment with activin or FS did not consistently affect cell growth. Measurement of serum activin A in a subset of subjects and in 27 additional subjects with epithelial ovarian carcinoma (n = 33) revealed preoperative activin A levels > 3 SD above the mean for pre- and postmenopausal women in 13/33 (39%) subjects. We conclude that in epithelial ovarian cancer: 1) beta A subunit mRNA is expressed, 2) activin protein is secreted more frequently than inhibin and in greater quantities than FS, 3) beta A subunit mRNA expression is greater in neoplastic and normal epithelium in culture than in the primary tissue, 4) the majority of tumors in culture do not respond to activin or FS treatment with proliferation, and 5) serum activin levels may reflect tumor secretion in some patients. Thus, activin A appears to be available as an autocrine/paracrine factor in epithelial ovarian tumors and may contribute to circulating levels, but its role in tumorigenesis has yet to be defined.


Subject(s)
Carcinoma/chemistry , Glycoproteins/analysis , Inhibins/analysis , Ovarian Neoplasms/chemistry , Activins , Adenocarcinoma, Clear Cell/chemistry , Adenocarcinoma, Mucinous/chemistry , Adult , Aged , Carcinoma/metabolism , Carcinoma/pathology , Cell Division , Cystadenocarcinoma, Serous/chemistry , Epithelium/chemistry , Female , Follistatin , Gene Expression , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Inhibins/genetics , Inhibins/metabolism , Middle Aged , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , RNA, Messenger/analysis
13.
Gynecol Oncol ; 65(3): 512-6, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9190984

ABSTRACT

Inhibin is an ovarian protein previously shown, using a nonspecific assay, to be elevated in serum of women with ovarian cancer. However, inhibin is secreted in multiple biochemical forms, including dimeric inhibin A and B and alpha inhibin precursors (pro-alphaC), each of which can now be specifically measured. We have examined the secretion of inhibin B and pro-alphaC inhibin in serum from women with epithelial ovarian cancer (EOC) for the first time, and have compared these analytes to inhibin A and total inhibin (inhibin A + B + pro-alphaC) as potential serum markers for EOC in postmenopausal women. Of all the immunoreactive inhibin proteins studied, the best serum marker was pro-alphaC, with 22% of women with EOC having levels that exceeded the range of values in women without EOC. Since CA 125 and pro-alphaC levels were not significantly correlated, combination of these markers resulted in 87% of EOC cases having elevated preoperative serum levels, a 9% increase over CA 125 alone. These data suggest that alpha inhibin secretion, especially pro-alphaC, may be useful in addition to CA 125 as a serum marker for EOC in postmenopausal women.


Subject(s)
Inhibins/blood , Ovarian Neoplasms/blood , Aged , Aged, 80 and over , Female , Humans , Middle Aged , Postmenopause
14.
Endocrinology ; 137(12): 5476-83, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8940374

ABSTRACT

LNCaP cells, derived from an androgen-sensitive cell line widely employed as an in vitro model of human prostate cancer, have been shown to express activin receptors. Activin is a local regulator of cellular growth, appears to play a key role in mesoderm induction and differentiation during development, and has been implicated in gonadal tumorigenesis. Follistatin, a monomeric glycoprotein that specifically binds and neutralizes activin, is often coexpressed with activin and, thus, modulates the autocrine/paracrine biological activity of this potent growth factor. We tested the hypothesis that LNCaP growth is modulated by the activin/follistatin system. Recombinant human activin A inhibited [3H]thymidine incorporation in a dose-dependent fashion with an ED50 of approximately 0.43 +/- 0.3 nM. Activin (0.1-3 nM) also inhibited dihydrotestosterone (DHT)-stimulated [3H]thymidine incorporation in LNCaP cells. Similarly, recombinant human inhibin A inhibited LNCaP proliferation, but was only 1/100th as potent as activin. Furthermore, activin (3 nM) induced a 3-fold increase in the extent of labeling of low mol wt DNA fragments typical of apoptosis. Activin-induced apoptosis was also indicated by an increase in the number of cells with reduced DNA content, as measured by flow cytometry of activin-treated cells. Both activin-mediated inhibition of cell proliferation and induction of apoptosis could be completely blocked by recombinant human follistatin. Based upon these results using an in vitro model, we speculate that activin functions locally to oppose androgen-driven cell proliferation and, thus, is a key factor controlling prostate growth. Reduced activin biosynthesis, increased follistatin secretion, or signaling defects in the activin receptor system should be further investigated in future studies as potential mechanisms underlying enhanced androgen-independent growth of human prostate cancer cells.


Subject(s)
Androgens/pharmacology , Apoptosis , Inhibins/pharmacology , Prostatic Neoplasms/pathology , Activin Receptors , Activins , Cell Division/drug effects , Cell Separation , DNA Fragmentation , Flow Cytometry , Humans , Male , Prostatic Neoplasms/metabolism , Receptors, Growth Factor/metabolism , Tumor Cells, Cultured/drug effects
15.
Fertil Steril ; 66(3): 425-9, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8751742

ABSTRACT

OBJECTIVE: To determine if embryos can modulate steroid hormone production by luteinized granulosa cells. DESIGN: Granulosa cells obtained from follicular aspirates were cultured alone or in the presence of a two-pronuclear zygote. The production of E2 and P by these cultures was evaluated by RIA. SETTING: In Vitro Fertilization Unit in an academic research environment. PATIENTS: Sixteen women undergoing IVF. INTERVENTIONS: Standard IVF-ET treatment cycle using leuprolide acetate for pituitary desensitization before hMG or urofollitropin for ovarian stimulation. MAIN OUTCOME MEASURES: Estradiol and P concentration in culture media of luteinized granulosa cells alone or granulosa cells cocultured with a two-pronuclear embryo. RESULTS: Both E2 and P production by luteinized granulosa cells was reduced when cultured in the presence of an embryo. CONCLUSIONS: Human embryos secrete a factor that regulates granulosa cell steroidogenesis.


Subject(s)
Estrogens/biosynthesis , Granulosa Cells/metabolism , Progesterone/biosynthesis , Zygote/physiology , Adult , Antineoplastic Agents, Hormonal/pharmacology , Coculture Techniques/methods , Culture Media/chemistry , Estrogens/analysis , Female , Fertilization in Vitro/methods , Granulosa Cells/cytology , Humans , Leuprolide/pharmacology , Progesterone/analysis , Radioimmunoassay , Zygote/cytology
16.
Endocrinology ; 137(9): 3667-73, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8756531

ABSTRACT

The FSH beta gene is stimulated by low frequency pulses of GnRH, but is unaffected or suppressed when GnRH is applied at higher frequencies or continuously. The current studies explored the hypothesis that GnRH frequency-dependent regulation of FSH beta may be mediated by pituitary expression of activin, which stimulates FSH beta messenger RNA (mRNA), and follistatin, which blocks activin. Using a system of perifused male rat pituitary cells, a reciprocal relationship was observed between FSH beta and follistatin mRNAs in response to different patterns of GnRH treatment. Pulses of GnRH (5 min; 10 nM) applied every 60 min stimulated FSH beta mRNA 14.0-fold with no change in follistatin mRNA. Pulses of GnRH applied every 30 and 15 min elicited stepwise increases in follistatin mRNA and decreases in FSH beta mRNA, and continuous GnRH stimulated follistatin mRNA 4.1-fold, with no significant increase in FSH beta mRNA. Stimulation of FSH beta mRNA by hourly GnRH pulses (3.7-fold) was blocked in the presence of 30 ng/ml recombinant follistatin (0.8-fold), suggesting that GnRH stimulation of FSH beta mRNA requires endogenous activin. Treatment of plated pituitary cells with continuous GnRH for 24 h confirmed that secretion of follistatin protein rises (1.5-fold) coincident with follistatin mRNA (1.7-fold) under conditions that suppress FSH beta mRNA (9% of the control value). When male rats were infused through arterial cannulas for 6 h with continuous GnRH (100 nM) or recombinant follistatin (5 micrograms/h), continuous GnRH suppressed FSH beta mRNA levels to 50% of the control value, and follistatin decreased expression to 61% of the control value. We conclude that GnRH stimulation of FSH beta mRNA is activin dependent, and pituitary follistatin production is a major pathway by which higher GnRH pulse frequencies suppress FSH beta mRNA. Changes in activin or follistatin tone, therefore, provide a mechanism by which LH and FSH can be differentially regulated by GnRH in a variety of physiological and pathophysiological conditions.


Subject(s)
Follicle Stimulating Hormone/genetics , Gene Expression Regulation , Glycoproteins/physiology , Gonadotropin-Releasing Hormone/physiology , Inhibins/physiology , Activins , Animals , Follicle Stimulating Hormone/metabolism , Follistatin , Glycoproteins/genetics , Glycoproteins/metabolism , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Gonadotropin-Releasing Hormone/pharmacology , Male , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins , Time Factors
18.
J Clin Endocrinol Metab ; 81(4): 1434-41, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8636347

ABSTRACT

The follistatin/activin/inhibin system increasingly appears to have important growth and differentiating effects in a variety of cell types, including cancer. We have developed a two-site immunoradiometric assay for measurement of human follistatin using two monoclonal antibodies against recombinant human follistatin. This cloned protein donor assay is sensitive (0.5 ng/mL), specific for free human follistatin, and precise (<5% within assay coefficient of variation). Using this assay, native human follistatin could be measured in human pituitary extracts, follicular fluid, and granulosa-luteal cell-conditioned medium. To identify and characterize human follistatin secreted by ovarian cancer cells, we screened five human ovarian carcinoma cell lines currently available from the American Type Culture Collection (Rockville, MD). One of these, a cell line derived from a teratocarcinoma (designated PA-1, American Type Culture Collection, CRL1572), secreted large (3 microg/10(6) cells per 24 h) quantities of immunoreactive follistatin constituitively. Increasing volumes of conditioned medium from these cultured cells generated response curves parallel to those of recombinant human follistatin 288 reference protein, human follicular fluid, or culture medium from human granulosa-luteal cells. Secretion of follistatin by PA-1 cells was time and cell-number dependent with 297.9 +/- 15.2, 654 +/- 29.8, and 940 +/- 49.1 ng follistatin secreted over 24 h by 1 x 10(5), 2 x 10(5), and 3 x 10(5) cells, respectively. Western and ligand blot analysis revealed that the immunoreactive follistatin secreted by PA-1 cells and isolated by sulfate-cellufine chromatography was identical to the molecular weight variants (32,000 and 35,000 Mr) of recombinant human follistatin 288. PA-1 cell-conditioned medium suppressed basal secretion of FSH by cultured rat anterior pituitary cells in a dose-dependent fashion. This follistatin bioactivity was completely removed by adsorption with either solid-phase monoclonal antifollistatin or a dextran-sulfate chromatography gel. Because activin suppressed the proliferation of PA-1 cells, secretion of bioactive follistatin may represent an autocrine mechanism opposing activin to maintain the rapid growth rate of PA-1 cells. These observations demonstrate that the ovarian teratocarcinoma cell line, PA-1, secretes considerable amounts of human follistatin that is biologically active, capable of binding human activin, and antigenically similar to recombinant human follistatin 288. The monoclonal antibodies and two-site assay reported herein should be useful in assessing the regulation of follistatin secretion and as a diagnostic tool, especially if follistatin measurements prove to be a marker for some ovarian cancers.


Subject(s)
Glycoproteins/analysis , Glycoproteins/metabolism , Ovarian Neoplasms/metabolism , Teratocarcinoma/metabolism , Activins , Animals , Antibodies, Monoclonal , Antibody Specificity , Biological Assay , Blotting, Western/methods , Cell Division/drug effects , Cell Line , Cells, Cultured , Corpus Luteum/cytology , Cross Reactions , Culture Media, Conditioned , Female , Follicular Fluid/chemistry , Follistatin , Glycoproteins/pharmacology , Granulosa Cells/cytology , Humans , Immunoradiometric Assay/methods , Inhibins/pharmacology , Male , Molecular Weight , Pituitary Gland/chemistry , Pituitary Gland, Anterior/drug effects , Pituitary Gland, Anterior/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/analysis , Recombinant Proteins/pharmacology , Reference Values , Reproducibility of Results , Sensitivity and Specificity , Tumor Cells, Cultured
19.
Fertil Steril ; 65(3): 667-9, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8774307

ABSTRACT

OBJECTIVE: To determine if serum inhibin concentrations are lower in ectopic (EP) versus intrauterine pregnancies (IUPs) that are conceived spontaneously. DESIGN: Case-control study. SETTING: Academic clinical practice. PATIENTS: Serum samples were obtained from 19 women who had EP confirmed at surgery and by pathology. For comparison, serum samples were collected from 24 women of similar chronological and gestational age with sonographic evidence of an IUP. MAIN OUTCOME MEASURE: Serum dimeric inhibin-A, total inhibin, P, and hCG. RESULTS: Serum total and dimeric inhibin concentrations in women with EP were < 60% of the concentrations for women with single IUPs. Total inhibin, but not dimeric inhibin-A, was elevated in maternal serum before week 8 of gestation relative to normal menstrual cycle levels. CONCLUSIONS: Serum inhibin concentrations are lower in EP as compared with IUPs that are spontaneously conceived and the relative amounts of dimeric inhibin-A, B, and alpha inhibin subunit in maternal serum may change throughout gestation.


Subject(s)
Inhibins/blood , Pregnancy, Ectopic/blood , Pregnancy/blood , Case-Control Studies , Chorionic Gonadotropin/blood , Female , Humans , Inhibins/chemistry , Osmolar Concentration , Progesterone/blood , Reference Values
20.
Endocrinology ; 137(2): 486-94, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8593793

ABSTRACT

Although ovarian cancer is the most common gynecological malignancy with a relatively poor 5-yr survival record, the mechanism(s) by which these tumors arise is not well understood. A role for inhibins and activins in regulating this transformation is suggested by the detection of circulating alpha or dimeric inhibin in some patients with ovarian cancer and by the alpha inhibin knockout mouse, in which development of gonadal tumors in 100% of homozygotes is associated with greatly elevated activin levels. To develop diagnostic tools with greater specificity for ovarian cancers, the present study was targeted at characterizing the biosynthetic capacity of the epithelial ovarian cancer cell lines from the American Type Culture Collection with respect to inhibin, activin, the related activin-binding protein follistatin (FS), and activin receptor type II. In addition, the functional capacity of this system was investigated by examining the ability of activin and FS to modulate cellular proliferation. All six cell lines contained abundant messenger RNA (mRNA) for activin receptor type II, but no inhibin alpha-subunit mRNA was detected in any cell line. Two cell lines contained mRNA for activin beta B-subunit (CaOV4 and SKOV3), one cell line contained beta A-subunit mRNA (SW626), and one cell line contained both (ES2); the latter also contained FS mRNA. FS mRNA was detected in another cell line (PA-1) that contained no detectable activin beta-subunit mRNA. Finally, one cell line (CaOV3) contained neither beta-subunit nor FS mRNA. Protein secretion was also examined. Consistent with the mRNA studies, the two cell lines containing FS mRNA secreted FS (PA-1 and ES2 cells), whereas three of the remaining lines secreted activin (A or B). In the cell line containing neither FS nor beta-subunit mRNA, no FS or activin could be detected. Finally, none of the cell lines secreted detectable immunoreactive inhibin. The effects of exogenous activin and FS on cellular proliferation were examined in these cell lines. No response was detected in the two cell lines that secreted FS (PA-1 and ES2). For the four cell lines not synthesizing FS, treatment with activin (1-100 ng/ml) resulted in an increase, whereas FS treatment (1-100 ng/ml) resulted in a decrease in cellular proliferation, as determined by [3H]thymidine incorporation. The response to activin correlated negatively with endogenous activin production, suggesting that autocrine activin production may be involved with cell proliferation. The differential expression and production of inhibin/activin subunits, activin receptors, and follistatin as well as the range of responses to exogenous activin among six ovarian epithelial cancer cell lines suggest that this family of hormones may be important in regulating cell proliferation in the ovary. Whether primary tumors have the same profile and the degree to which these results can be generalized to additional forms of ovarian cancer remain to be determined.


Subject(s)
Glycoproteins/metabolism , Inhibins/metabolism , Ovarian Neoplasms/metabolism , Receptors, Growth Factor/metabolism , Activin Receptors , Activins , Base Sequence , Cell Division/drug effects , Female , Follistatin , Glycoproteins/genetics , Homeostasis , Humans , Inhibins/genetics , Molecular Sequence Data , Oligonucleotide Probes/genetics , Ovarian Neoplasms/pathology , RNA, Messenger/metabolism , Receptors, Growth Factor/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...