Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
Add more filters










Publication year range
1.
Endocrinology ; 163(3)2022 03 01.
Article in English | MEDLINE | ID: mdl-35022746

ABSTRACT

Testicular-derived inhibin B (α/ß B dimers) acts in an endocrine manner to suppress pituitary production of follicle-stimulating hormone (FSH), by blocking the actions of activins (ß A/B/ß A/B dimers). Previously, we identified a homozygous genetic variant (c.1079T>C:p.Met360Thr) arising from uniparental disomy of chromosome 2 in the INHBB gene (ß B-subunit of inhibin B and activin B) in a man suffering from infertility (azoospermia). In this study, we aimed to test the causality of the p.Met360Thr variant in INHBB and testis function. Here, we used CRISPR/Cas9 technology to generate InhbbM364T/M364T mice, where mouse INHBB p.Met364 corresponds with human p.Met360. Surprisingly, we found that the testes of male InhbbM364T/M364T mutant mice were significantly larger compared with those of aged-matched wildtype littermates at 12 and 24 weeks of age. This was attributed to a significant increase in Sertoli cell and round spermatid number and, consequently, seminiferous tubule area in InhbbM364T/M364T males compared to wildtype males. Despite this testis phenotype, male InhbbM364T/M364T mutant mice retained normal fertility. Serum hormone analyses, however, indicated that the InhbbM364T variant resulted in reduced circulating levels of activin B but did not affect FSH production. We also examined the effect of this p.Met360Thr and an additional INHBB variant (c.314C>T: p.Thr105Met) found in another infertile man on inhibin B and activin B in vitro biosynthesis. We found that both INHBB variants resulted in a significant disruption to activin B in vitro biosynthesis. Together, this analysis supports that INHBB variants that limit activin B production have consequences for testis composition in males.


Subject(s)
Infertility, Male/genetics , Inhibin-beta Subunits/genetics , Inhibin-beta Subunits/physiology , Mutation , Sperm Count , Testis/physiopathology , Activins/biosynthesis , Activins/genetics , Animals , Azoospermia/genetics , CRISPR-Associated Protein 9 , Follicle Stimulating Hormone/metabolism , Humans , Infertility, Male/physiopathology , Inhibins/biosynthesis , Inhibins/genetics , Male , Mice , Mice, Inbred C57BL , Sertoli Cells , Spermatogenesis/genetics , Spermatogonia , Testis/chemistry , Testis/cytology
2.
Mol Biol Rep ; 46(2): 1603-1609, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30680594

ABSTRACT

Activin E, a member of the TGF-ß super family, is a protein dimer of mature inhibin ßE subunits. Recently, it is reported that hepatic activin E may act as a hepatokine that alter whole body energy/glucose metabolism in human. However, orthologues of the activin E gene have yet to be identified in lower vertebrates, including fish. Here, we cloned the medaka (Oryzias latipes) activin E cDNA from liver. Among all the mammalian inhibin ß subunits, the mature medaka activin E amino acid sequence shares the highest homology with mammalian activin E. Recombinant expression studies suggest that medaka activin E, the disulfide-bound mature form of mature inhibin ßE subunits, may exert its effects in a way similar to that in mammals. Although activin E mRNA is predominantly expressed in liver in mammals, it is ubiquitously expressed in medaka tissues. Since expression in the liver was enhanced after a high fat diet, medaka activin E may be associated with energy/glucose metabolism, as shown in mice and human.


Subject(s)
Inhibin-beta Subunits/metabolism , Inhibin-beta Subunits/physiology , Oryzias/genetics , Activins/metabolism , Activins/physiology , Amino Acid Sequence , Animals , DNA, Complementary/metabolism , Inhibins/genetics , Inhibins/metabolism , Liver/metabolism , Oryzias/metabolism , RNA, Messenger/metabolism , Transforming Growth Factor beta/metabolism
3.
Reproduction ; 155(1): 15-23, 2018 01.
Article in English | MEDLINE | ID: mdl-29030492

ABSTRACT

Activin A regulates testicular and epididymal development, but the role of activin B in the epididymis and vas deferens is unknown. Mouse models with reduced activin A (Inhba+/- and InhbaBK/+), or its complete absence (InhbaBK/BK), were investigated to identify specific roles of activins in the male reproductive tract. In 8-week-old Inhba+/- mice, serum activin A decreased by 70%, with a 50% reduction of gene expression and protein in the testis, epididymis and vas deferens. Activin B and the activin-binding protein, follistatin, were similar to wild-type. Testis weights were slightly reduced in Inhba+/- mice, but the epididymis and vas deferens were normal, while the mice were fertile. Activin A was decreased by 70% in the serum, testis, epididymis and vas deferens of InhbaBK/+ mice and was undetectable in InhbaBK/BK mice, but activin B and follistatin levels were similar to wild-type. In 6-week-old InhbaBK/BK mice, testis weights were 60% lower and epididymal weights were 50% lower than in either InhbaBK/+ or wild-type mice. The cauda epididymal epithelium showed infoldings and less intra-luminal sperm, similar to 3.5-week-old wild-type mice, but at 8 weeks, no structural differences in the testis or epididymis were noted between InhbaBK/BK and wild-type mice. Thus, Inhbb can compensate for Inhba in regulating epididymal morphology, although testis and epididymal maturation is delayed in mice lacking Inhba Crucially, reduction or absence of activin A, at least in the presence of normal activin B levels, does not lead to major defects in the adult epididymis or vas deferens.


Subject(s)
Epididymis/metabolism , Gene Expression Regulation , Inhibin-beta Subunits/physiology , Vas Deferens/metabolism , Animals , Male , Mice , Mice, Knockout
4.
J Clin Endocrinol Metab ; 99(10): E1922-32, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25062451

ABSTRACT

CONTEXT: Exerting a broad range of biological effects in various tissues, activins are homo- or heterodimers of activin/inhibin ß-subunits (ßA, ßB, ßC, and ßE in humans). Although activins A (ßAßA), B (ßBßB), AB (ßAßB), and AC (ßAßC) have been demonstrated in the female reproductive system, little is known about their individual functions in the ovary. OBJECTIVE: To investigate the biological roles and activities of activins in regulating steroidogenesis in human granulosa cells. DESIGN: Human granulosa-lutein cells obtained from 32 patients undergoing in vitro fertilization were used to investigate the effects of activin A, B, AB, and AC on the expression of steroidogenic enzymes and steroid production. SETTING: An academic research center. MAIN OUTCOME MEASURES: mRNA and protein levels were examined by reverse transcription quantitative real-time PCR and Western blot analysis, respectively. The production of estradiol and progesterone was measured by enzyme immunoassay. RESULTS: P450 aromatase, FSH receptor, and estradiol levels were increased, whereas steroidogenic acute regulatory protein (StAR), LH receptor, and progesterone levels were decreased after treatment with activin A, B, and AB, but not activin AC. FSH or LH induced the production of aromatase/estradiol and StAR/progesterone; however, pretreatment with activin A, B, or AB enhanced the effects of gonadotropins on aromatase/estradiol, but suppressed their effects on StAR/progesterone. Treatment with activin A, B, or AB induced the phosphorylation of SMA- and MAD-related proteins (SMAD2 and 3), whereas activin AC had no such effects. Furthermore, co-culture of activin AC (1-100 ng/mL) with activin A (25 ng/mL) did not alter the effects of activin A on P450 aromatase or StAR mRNA levels. CONCLUSION: Activin A, B, and AB have similar effects on steroidogenesis in human granulosa cells. In contrast, activin AC is not biologically active and does not act as a competitive antagonist.


Subject(s)
Activins/physiology , Inhibin-beta Subunits/physiology , Luteal Cells/physiology , Activins/genetics , Activins/pharmacology , Aromatase/metabolism , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Activation/physiology , Estradiol/biosynthesis , Female , Humans , Inhibin-beta Subunits/genetics , Inhibin-beta Subunits/pharmacology , Luteal Cells/cytology , Luteal Cells/drug effects , Ovary/cytology , Phosphoproteins/metabolism , Progesterone/biosynthesis , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology
5.
Anim Reprod Sci ; 148(3-4): 97-108, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25001504

ABSTRACT

Cystic ovarian disease (COD) is an important cause of infertility in dairy cattle. Although many researchers have focused their work on the endocrine changes related to this disease, evidence indicates that intraovarian components play an important role in follicular persistence. Activin, inhibin, and follistatin participate as intraovarian regulatory molecules involved in follicular cell proliferation, differentiation, steroidogenesis, oocyte maturation, and corpus luteum function. Given the importance of these factors in folliculogenesis, we examined the expression and immunolocalization of activin/inhibin ßA-subunit, inhibin α-subunit, and follistatin in the ovaries of healthy estrus-synchronized cows and in those of cows with spontaneous or adrenocorticotropic hormone (ACTH)-induced COD. We also studied inhibin B (α ßB) levels in serum and follicular fluid. We found an increased expression of the ßA-subunit of activin A/inhibin A, the α-subunit of inhibin, and follistatin in granulosa cells of spontaneous follicular cysts by immunohistochemistry, and decreased concentrations of inhibin B (α ßB) in the follicular fluid of spontaneous follicular cysts. These results, together with those previously obtained, indicate that the expression of the components of the activin-inhibin-follistatin system is altered. This could lead to multiple alterations in important functions in the ovary like the balance between pro- and anti-apoptotic factors, follicular proliferation/apoptosis, and steroidogenesis, which may contribute to the follicular persistence and endocrine changes found in cattle with COD.


Subject(s)
Cattle Diseases/etiology , Follistatin/physiology , Inhibin-beta Subunits/physiology , Inhibins/physiology , Ovarian Cysts/etiology , Adrenocorticotropic Hormone , Animals , Blood/metabolism , Case-Control Studies , Cattle , Cattle Diseases/diagnostic imaging , Cattle Diseases/metabolism , Female , Follicular Fluid/metabolism , Follistatin/metabolism , Inhibin-beta Subunits/metabolism , Inhibins/metabolism , Ovarian Cysts/diagnostic imaging , Ovarian Cysts/metabolism , Ovarian Cysts/veterinary , Protein Subunits , Ultrasonography
6.
Molecules ; 18(10): 12977-86, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24141247

ABSTRACT

Activin A (ActA), a member of transforming growth factor-beta (TGF-b) super- family, affects many cellular processes, including ischemic stroke. Though the neuroprotective effects of exogenous ActA on oxygen-glucose deprivation (OGD) injury have already been reported by us, the endogenous role of ActA remains poorly understood. To further define the role and mechanism of endogenous ActA and its signaling in response to acute ischemic damage, we used an OGD model in PC12 cells to simulate ischemic injury on neurons in vitro. Cells were pre-treated by monoclonal antibody against activin receptor type IIA (ActRII-Ab). We found that ActRII-Ab augments ischemic injury in PC12 cells. Further, the extracellular secretion of ActA as well as phosphorylation of smad3 in PC12 cells was also up-regulated by OGD, but suppressed by ActRII-Ab. Taken together, our results show that ActRII-Ab may augment ischemic injury via blocking of transmembrane signal transduction of ActA, which confirmed the existence of endogenous neuroprotective effects derived from the ActA/Smads pathway. ActRIIA plays an important role in transferring neuronal protective signals inside. It is highly possible that ActA transmembrance signaling is a part of the positive feed-back loop for extracellular ActA secretion.


Subject(s)
Inhibin-beta Subunits/physiology , Signal Transduction , Smad3 Protein/metabolism , Activin Receptors, Type II/antagonists & inhibitors , Activin Receptors, Type II/metabolism , Animals , Cell Hypoxia , Cell Survival , Glucose/deficiency , Hypoxia-Ischemia, Brain/metabolism , PC12 Cells , Phosphorylation , Protein Processing, Post-Translational , Rats
7.
Anim Sci J ; 82(1): 57-61, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21269360

ABSTRACT

The inhibin ß(B) (INHBB) gene was studied as a candidate gene for the prolificacy of Small Tail Han and Hu sheep. According to the sequence of exon 1 and 2 of bovine INHBB gene, six pairs of primers were designed to detect single nucleotide polymorphisms of exon 1 and 2 of INHBB gene in both high (Small Tail Han and Hu sheep) and low prolificacy breeds (Dorset, Texel and German Mutton Merino sheep) by polymerase chain reaction-single strand conformation polymorphism (PCR-SSCP). Three pairs of primers (primers 1-1, 1-2 and 1-3) were used to amplify the exon 1, and others (primers 2-1, 2-2 and 2-3) to the exon 2. Only the products amplified by primer 2-3 displayed polymorphism. For primer 2-3, three genotypes (AA, AB and BB) were detected in Hu sheep and only AA genotype in other breeds. In Hu sheep, frequency of AA, AB and BB genotypes was 0.636, 0.046 and 0.318, respectively. Sequencing revealed 276A > G mutation (based on the amplification region of primer 2-3) which did not cause any amino acid change because it lay in the 3' untranslated region. The ewes with genotype BB had 0.58 (P < 0.01) lambs more than those with AA in Hu sheep.


Subject(s)
Inhibin-beta Subunits/genetics , Inhibin-beta Subunits/physiology , Litter Size/genetics , Polymorphism, Single Nucleotide , Sheep/genetics , Sheep/physiology , Animals , Exons/genetics , Female , Gene Frequency , Genotype , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational
8.
Biochem Biophys Res Commun ; 395(3): 373-6, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20382119

ABSTRACT

Activin B, consisting of two inhibin betaB (INHBB) subunits, is a hormone known to affect gonadal function, reproduction and fetal development. We have reported that INHBB and activin B receptors are highly expressed in adipocytes suggesting that activin B may have local effects in adipose tissue. In this study, we investigate the effect of activin B on lipolysis, measured as release of non-esterified fatty acids and free glycerol. Recombinant activin B decreased lipolysis in a concentration-dependent manner and increased intracellular triglyceride content in 3T3-L1 adipocytes. siRNA-mediated knock-down of INHBB expression increased lipolysis, and this effect was abolished by addition of recombinant activin B. In line with its inhibitory effect on lipolysis, activin B caused a down regulation of the expression of adipose triglyceride lipase and hormone sensitive lipase, key genes involved in lipolysis. In summary, we suggest that activin B is a novel adipokine that inhibits lipolysis in a paracrine or autocrine manner.


Subject(s)
Activins/physiology , Adipocytes/physiology , Lipolysis/physiology , 3T3-L1 Cells , Activins/genetics , Activins/pharmacology , Adipocytes/drug effects , Animals , Fatty Acids, Nonesterified/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Glycerol/metabolism , Inhibin-beta Subunits/genetics , Inhibin-beta Subunits/physiology , Lipolysis/drug effects , Lipolysis/genetics , Mice , RNA, Small Interfering/genetics , Triglycerides/metabolism
9.
Biochem Biophys Res Commun ; 385(3): 319-23, 2009 Jul 31.
Article in English | MEDLINE | ID: mdl-19463785

ABSTRACT

To study the function of activin E, a TGF-beta superfamily member, in the regulation of affective behavior, we investigated the behavior of transgenic mice overexpressing activin E (TgActbetaE mice). Male TgActbetaE mice showed aggressive behavior in resident-intruder tests. In elevated plus-maze tests, the percentage of open arm entries was significantly increased in female TgActbetaE mice compared with that in wild-type mice. Furthermore, female TgActbetaE mice stayed in the central area for a significantly longer time than wild-type mice in open field tests. These results indicated that TgActbetaE mice had less anxiety-like behavior. The number of restraint-stress-evoked c-Fos-positive cells in the hypothalamic paraventricular nucleus in TgActbetaE mice was significantly decreased compared with that in wild-type mice. This suggests that synthesis of corticotrophin-releasing hormone induced by stress was decreased in TgActbetaE mice. Taking these results together, activin E may act as a regulator of the hypothalamic-pituitary-adrenal axis.


Subject(s)
Aggression , Anxiety/genetics , Hypothalamo-Hypophyseal System/physiology , Inhibin-beta Subunits/physiology , Pituitary-Adrenal System/physiology , Animals , Anxiety/psychology , Female , Inhibin-beta Subunits/genetics , Male , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-fos/metabolism
10.
Endocrinology ; 150(8): 3521-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19389832

ABSTRACT

Activin-betaA and activin-betaB (encoded by Inhba and Inhbb genes, respectively) are closely related TGF-beta superfamily members that participate in a variety of biological processes. We previously generated mice with an insertion allele at the Inhba locus, Inhba(BK). In this allele, the sequence encoding the Inhba mature domain is replaced with that of Inhbb, rendering the gene product functionally hypomorphic. Homozygous (Inhba(BK/BK)) and hemizygous (Inhba(BK/-)) mice are smaller and leaner than their wild-type littermates, and many tissues are disproportionately small relative to total body weight. To determine the mechanisms that contribute to these phenomena, we investigated the metabolic consequences of the mutation. Although the growth of Inhba(BK) mice is improved by providing a calorie-rich diet, diet-induced obesity, fatty liver, and insulin resistance (hallmarks of chronic caloric excess) do not develop, despite greater caloric intake than wild-type controls. Physiological, molecular, and biochemical analyses all revealed characteristics that are commonly associated with increased mitochondrial energy metabolism, with a corresponding up-regulation of several genes that reflect enhanced mitochondrial biogenesis and function. Oxygen consumption, an indirect measure of the metabolic rate, was markedly increased in Inhba(BK/BK) mice, and polarographic analysis of liver mitochondria revealed an increase in ADP-independent oxygen consumption, consistent with constitutive uncoupling of the inner mitochondrial membrane. These findings establish a functional relationship between activin signaling and mitochondrial energy metabolism and further support the rationale to target this signaling pathway for the medical treatment of cachexia, obesity, and diabetes.


Subject(s)
Body Composition/genetics , Energy Metabolism/genetics , Inhibin-beta Subunits/physiology , Animals , Blood Glucose/drug effects , Blood Glucose/genetics , Body Weight/genetics , Calorimetry, Indirect , Dietary Fats/pharmacology , Energy Intake/genetics , Enzyme-Linked Immunosorbent Assay , Female , Glucose Tolerance Test , In Vitro Techniques , Inhibin-beta Subunits/genetics , Insulin Resistance/genetics , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Male , Mice , Mice, Mutant Strains , Organ Size/genetics , Oxygen Consumption , Radioimmunoassay , Reverse Transcriptase Polymerase Chain Reaction
11.
Nephrology (Carlton) ; 14(3): 311-20, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19298640

ABSTRACT

AIM: The effect of activin A on tubulointerstitial fibrosis in diabetic nephropathy (DN) using streptozotocin (STZ)-induced diabetic rats and high glucose-cultured HK-2 cells was investigated. METHODS: Male Wistar rats were randomized into a normal control group (NC) and diabetes mellitus group (DM). Diabetes was induced by i.p. injection of STZ. Six rats were respectively killed 4, 8, 12 and 16 weeks after model establishment in each group. The changes of kidney weight/bodyweight (KW/BW), urine albumin excretion rate (AER) and creatinine clearance rate (Ccr) were determined. The morphology of tubulointerstitium was observed by light microscopy. Further biochemical analysis was provided using immunohistochemistry and real-time polymerase chain reaction. The different parameters in high glucose-cultured HK-2 cells were monitored by western blotting or enzyme-linked immunosorbent assay (ELISA) and the intervention of rh-follistatin on them was investigated. RESULTS: Compared with the NC group, there was marked enlargement in the levels of KW/BW, AER, Ccr and interstitial fibrosis index, and the production of P-Smad2/3 and fibronectin in the DM group from 8 to 16 weeks. Activin betaA, mainly located in tubular epithelial cells, was significantly higher in the DM group than that in the NC group throughout the study periods. Follistatin was abundant in the NC group, but was diminished gradually in the DM group. High glucose may facilitate the synthesis of activin betaA, transforming growth factor (TGF)-beta, P-Smad2/3 and fibronectin in HK-2 cells while rh-follistatin inhibited them except TGF-beta. CONCLUSION: Activin A is involved in tubulointerstitial fibrosis in DN by inducing the production of fibronectin through Smad signal pathway.


Subject(s)
Diabetic Nephropathies/pathology , Inhibin-beta Subunits/physiology , Kidney Tubules/pathology , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/complications , Fibronectins/genetics , Fibrosis , Follistatin/analysis , Follistatin/genetics , Immunohistochemistry , Male , Rats , Rats, Wistar , Smad2 Protein/physiology , Smad3 Protein/physiology
12.
J Thorac Oncol ; 4(4): 455-62, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19240652

ABSTRACT

INTRODUCTION: The expression, mechanisms of regulation, and functional impact of activin (INHBA) in esophageal adenocarcinoma (EAC) have not been fully defined. METHODS: INHBA expression was examined in 46 esophageal samples (nine Barrett's metaplasia (BM); seven BM/low-grade dysplasia; eight low-grade dysplasia; seven high-grade dysplasia; 15 EAC) using oligonucleotide microarrays and real-time reverse transcription-polymerase chain reaction (RT-PCR) and in 90 tissue samples (79 EAC; 8 dysplastic; 3 BM) using immunohistochemistry (IHC). The proliferation of EAC cell lines FLO and OE-33 was examined after treatment with exogenous activin. The proliferation of OE-33 was also examined after treatment with the activin inhibitor follistatin and INHBA-targeting siRNA. OE-33 and FLO cells were treated with 5-aza-2'deoxycytidine (5-AZA) and trichostatin A to investigate the role of epigenetic regulation in INHBA expression. RESULTS: Primary EACs expressed 5.7-times more INHBA mRNA than BM samples on oligonucleotide microarray. Transcript overexpression in EAC relative to BM was confirmed on real-time RT-PCR. IHC suggested higher INHBA protein expression in EAC (69.6%) than in the dysplastic (37.5%) and BM samples (33.3%). FLO and OE-33 treated with activin demonstrated increased proliferation, and OE-33 cells treated with follistatin and INHBA-targeting siRNA demonstrated reduced proliferation, relative to untreated controls. Treatment of FLO cells with trichostatin A and 5-AZA up-regulated INHBA mRNA and protein production by real time RT-PCR and IHC. CONCLUSIONS: INHBA is overexpressed in EAC relative to dysplastic and BM tissue. INHBA overexpression may promote cell proliferation and may be affected by promoter demethylation and histone acetylation in EAC cell lines.


Subject(s)
Adenocarcinoma/pathology , Epigenesis, Genetic , Esophageal Neoplasms/pathology , Inhibin-beta Subunits/physiology , Activins/pharmacology , Animals , Cattle , Cell Line, Tumor , Cell Proliferation/drug effects , Follistatin/pharmacology , Histone Acetyltransferases/genetics , Humans , Immunohistochemistry , Inhibin-beta Subunits/analysis , Inhibin-beta Subunits/antagonists & inhibitors , Inhibin-beta Subunits/genetics , RNA, Messenger/analysis , RNA, Small Interfering/genetics
13.
Endocrinology ; 150(4): 1904-11, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19022896

ABSTRACT

Abnormal uterine bleeding can severely affect the quality of life for women. After menstruation, the endometrium must adequately repair to limit and stop bleeding. Abnormal uterine bleeding may result from incorrect or inadequate endometrial repair after menstruation. Previous studies have shown an important contribution of activin to skin wound healing, with severely delayed wound repair observed in animals transgenically induced to overexpress activin's natural inhibitor, follistatin. Activin subunits have also been identified within human endometrium; however, their role in endometrial repair is unknown. We assessed the contribution of activin to endometrial repair after menses using a human in vitro cell wounding method and our well-characterized mouse model of endometrial breakdown and repair applied to mice overexpressing follistatin. Endometrial repair after menses is initiated with reepithelialization of the uterine surface. To mimic this repair, we utilized a human endometrial epithelial cell line (ECC-1) and demonstrated significant stimulation of wound closure after activin A administration, and attenuation of this response by addition of follistatin. Immunolocalization of activin subunits, betaA and betaB, in control endometrium from the mouse model demonstrated specific epithelial and stromal localization and some leukocyte staining (betaA) around sites of endometrial repair, suggestive of a role for activin in this process. Follistatin-overexpressing animals had significantly higher circulating follistatin levels than wild-type littermates. There was a significant delay in endometrial repair after breakdown in follistatin transgenic animals compared with control animals. This study demonstrates for the first time a functional role for activin in endometrial repair after menses.


Subject(s)
Activins/metabolism , Activins/physiology , Endometrium/metabolism , Menstruation/psychology , Activins/pharmacology , Animals , Cell Line , Cells, Cultured , Disease Models, Animal , Female , Follistatin/pharmacology , Humans , Immunohistochemistry , Inhibin-beta Subunits/genetics , Inhibin-beta Subunits/metabolism , Inhibin-beta Subunits/physiology , Male , Menstruation/metabolism , Mice , Mice, Transgenic , Wound Healing/drug effects , Wound Healing/physiology
14.
Differentiation ; 76(5): 443-53, 2008 May.
Article in English | MEDLINE | ID: mdl-18093225

ABSTRACT

The mechanisms controlling endodermal development during stem cell differentiation have been only partly elucidated, although previous studies have suggested the participation of fibroblast growth factor (FGF) and activin A in these processes. Shb is a Src homology 2 (SH2) domain-containing adapter protein that has been implicated in FGF receptor 1 (FGFR1) signaling. To study the putative crosstalk between activin A and Shb-dependent FGF signaling in the differentiation of endoderm from embryonic stem (ES) cells, embryoid bodies (EBs) derived from mouse ES cells overexpressing wild-type Shb or Shb with a mutated SH2 domain (R522K-Shb) were cultured in the presence of activin A. We show that expression of R522K-Shb results in up-regulation of FGFR1 and FGF2 in EBs. Addition of activin A to the cultures enhances the expression of endodermal genes primarily in EBs expressing mutant Shb. Inhibition of FGF signaling by the addition of the FGFR1 inhibitor SU5402 completely counteracts the synergistic effects of R522K-Shb and activin A. In conclusion, the present results suggest that expression of R522K-Shb enhances certain signaling pathways downstream of FGF and that an interplay between FGF and activin A participates in ES cell differentiation to endoderm.


Subject(s)
Endoderm/cytology , Fibroblast Growth Factor 2/physiology , Inhibin-beta Subunits/physiology , Proto-Oncogene Proteins/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured/cytology , Cells, Cultured/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endoderm/metabolism , Fibroblast Growth Factor 2/biosynthesis , Fibroblast Growth Factor 2/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental/physiology , Hepatocyte Nuclear Factors/biosynthesis , Hepatocyte Nuclear Factors/genetics , Inhibin-beta Subunits/genetics , Inhibin-beta Subunits/pharmacology , Mice , Mutation, Missense , Organoids/cytology , Organoids/metabolism , Point Mutation , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor Cross-Talk/physiology , Receptor, Fibroblast Growth Factor, Type 1/biosynthesis , Receptor, Fibroblast Growth Factor, Type 1/genetics , Recombinant Fusion Proteins/physiology , Signal Transduction/physiology , src Homology Domains/genetics
15.
Ann Surg Oncol ; 15(1): 96-103, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17909904

ABSTRACT

BACKGROUND: Expression of activin A is associated with lymph node metastasis and clinical stage in esophageal cancer. METHODS: To clarify the aggressive behavior of tumors with high activin A expression, we used the beta subunit of activin A to establish stable activin betaA (Act-betaA)-transfected carcinoma cells in two human esophageal carcinoma cell lines, KYSE110 and KYSE140. The biological behavior of these cells was compared with that in mock-transfected cells from the same cell lines. We focused our attention on cell growth and tumorigenesis, and proliferation and apoptosis. RESULTS: Both Act-betaA-transfected carcinoma cell lines showed a higher growth rate than the mock-transfected carcinoma cells. In an in vitro invasion assay and a xenograft analysis, the Act-betaA-transfected carcinoma cells showed far higher proliferation in vitro and a higher potency for tumorigenesis in vivo, respectively. Moreover, in an analysis of apoptosis via Fas stimulation, the Act-betaA-transfected carcinoma cells showed a higher tolerance to apoptosis compared with the mock-transfected carcinoma cells. Moreover, anti-activin-neutralizing antibody-treated squamous cell cancer cell lines inhibited their migration. CONCLUSIONS: Collectively, these data indicate that continuous high expression of activin A in esophageal carcinoma cells is not related to tumor suppression, but rather to tumor progression in vitro and in vivo. The inhibition of activin might be one of the methods to attenuate tumor aggressiveness.


Subject(s)
Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Inhibin-beta Subunits/physiology , Animals , Apoptosis/physiology , Blotting, Northern , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Movement , Cell Proliferation , Esophageal Neoplasms/genetics , Humans , Lymphatic Metastasis , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Smad Proteins/metabolism , Transfection , Tumor Cells, Cultured , fas Receptor/metabolism
16.
Mol Endocrinol ; 21(10): 2458-71, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17609433

ABSTRACT

Activins have diverse roles in multiple physiological processes including reproduction. Mutations and loss of heterozygosity at the human activin receptor ACVR1B and ACVR2 loci are observed in pituitary, pancreatic, and colorectal cancers. Functional studies support intraovarian roles for activins, although clarifying the in vivo roles has remained elusive due to the perinatal death of activin betaA knockout mice. To study the roles of activins in ovarian growth, differentiation, and cancer, a tissue-specific knockout system was designed to ablate ovarian production of activins. Mice lacking ovarian activin betaA were intercrossed to Inhbb homozygous null mice to produce double activin knockouts. Whereas ovarian betaA knockout females are subfertile, betaB/betaA double mutant females are infertile. Strikingly, the activin betaA and betaB/betaA-deficient ovaries contain increased numbers of functional corpora lutea but do not develop ovarian tumors. Microarray analysis of isolated granulosa cells identifies significant changes in expression for a number of genes with known reproductive roles, including Kitl, Taf4b, and Ghr, as well as loss of expression of the proto-oncogene, Myc. Thus, in contrast to the known tumor suppressor role of activins in some tissues, our data indicate that activin betaA and betaB function redundantly in a growth stimulatory pathway in the mammalian ovary.


Subject(s)
Fertility/genetics , Inhibin-beta Subunits/physiology , Ovary/metabolism , Animals , Carrier Proteins/genetics , Corpus Luteum/metabolism , Female , Gene Expression , Granulosa Cells/metabolism , Inhibin-beta Subunits/genetics , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Ovary/cytology , Proto-Oncogene Mas , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics
17.
Proc Natl Acad Sci U S A ; 104(27): 11322-7, 2007 Jul 03.
Article in English | MEDLINE | ID: mdl-17592132

ABSTRACT

Testis-derived testosterone has been recognized as the key factor for morphogenesis of the Wolffian duct, the precursor of several male reproductive tract structures. Evidence supports that testosterone is required for the maintenance of the Wolffian duct via its action on the mesenchyme. However, it remains uncertain how testosterone alone is able to facilitate formation of regionally specific structures such as the epididymis, vas deferens, and seminal vesicle from a straight Wolffian duct. In this study, we identified inhibin beta A (or Inhba) as a regional paracrine factor in mouse mesonephroi that controls coiling of the epithelium in the anterior Wolffian duct, the future epididymis. Inhba was expressed specifically in the mesenchyme of the anterior Wolffian duct at embryonic day 12.5 before the production of androgens. In the absence of Inhba, the epididymis failed to develop the characteristic coiling in the epithelium, which showed a dramatic decrease in proliferation. This loss of epididymal coiling did not result from testosterone deficiency, because testosterone production and parameters for testosterone action such as testis descent and anogenital distance remained normal. We further found that initial Inhba expression did not require testosterone as Inhba was also expressed in the anterior Wolffian duct of female embryos where no testosterone was produced. However, Inhba expression at later stages depended on testosterone. These results demonstrated that Inhba, a mesenchyme-specific gene, acts collectively with testosterone to facilitate epididymal coiling by stimulating epithelial proliferation.


Subject(s)
Epididymis/embryology , Inhibin-beta Subunits/physiology , Animals , Cell Proliferation , Inhibin-beta Subunits/deficiency , Inhibin-beta Subunits/genetics , Male , Mice , Mice, Knockout , Organ Culture Techniques , Testosterone/physiology , Wolffian Ducts/embryology
19.
J Reprod Dev ; 52(4): 487-95, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16627954

ABSTRACT

Activins, TGF-beta superfamily members, have multiple functions in a variety of cells and tissues. Additional activin beta subunit genes, betaC and betaE, have been identified in humans and rodents. To explore the role of activin betaC subunit, we generated recombinant human activin C using Chinese hamster ovary cells. Recombinant activin C from the conditioned medium was purified by consecutive hydrophobic, size-exclusion, and high performance liquid chromatography. SDS-PAGE and Western blot analysis of the purified protein revealed that activin C formed disulfide bridges. However, activin C had no effect on the proliferation of cultured liver cells. Furthermore, there were no significant differences in erythroid differentiation and follicle stimulating hormone secretion in vitro. It was also shown that immunoreactive bands indicated the hetrodimer of activin betaC, and inhibin alpha subunits were detected in the conditioned medium from the activin C-producing cells, which were stably transfected with inhibin alpha subunit cDNA. This suggests that activin betaC subunit may have been present and that it may exert its effect as inhibin C.


Subject(s)
Inhibin-beta Subunits/analysis , Inhibin-beta Subunits/physiology , Recombinant Proteins/analysis , Animals , CHO Cells , Cell Differentiation , Cricetinae , Cricetulus , Dimerization , Dose-Response Relationship, Drug , Follicle Stimulating Hormone/metabolism , Genetic Techniques , Humans , Inhibins/chemistry , K562 Cells , Liver/metabolism , Rats , Recombinant Proteins/chemistry
20.
J Mol Endocrinol ; 36(1): 201-20, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16461939

ABSTRACT

In rodents, activins stimulate immediate-early increases in pituitary follicle-stimulating hormone beta (Fshb) subunit transcription. Here, we investigated the underlying signaling mechanisms using the mouse gonadotrope cell line, LbetaT2. Activin A increased mouse Fshb-luciferase reporter activity within 4 h through a Smad-dependent signaling pathway. The ligand rapidly stimulated formation of SMAD2/3/4 complexes that could interact with a consensus palindromic Smad binding element (SBE) in the proximal Fshb promoter. SMAD over-expression potently stimulated transcription, with the combination of SMADs 2, 3 and 4 producing the greatest synergistic activation. A mutation in the SBE that abolished Smad binding greatly impaired the effects of acute (4 h) activin A treatment and SMAD over-expression on promoter activity, but did not abolish the effects of chronic (24 h) activin A exposure. Within activated SMAD complexes, SMADs 3 and 4 appeared to bind the SBE simultaneously and the binding of both was required for maximal transcriptional activation. Interestingly, the human FSHB promoter, which lacks the consensus SBE, was neither rapidly stimulated by activin A nor by over-expressed SMADs, but was activated by 24 h activin A. Addition of the SBE to the human promoter increased both SMAD2/3/4-sensitivity and acute regulation by activin A, though not to levels observed in mouse. We postulate that short reproductive cycles in female rodents, particularly the brief interval between the primary and secondary FSH surges of the estrous cycle, require the Fshb promoter in these animals to be particularly sensitive to the rapid, Smad-dependent actions of activins on transcription. The human FSHB promoter, in contrast, is chronically regulated by activins seemingly through a SMAD-independent pathway.


Subject(s)
Activins/physiology , Follicle Stimulating Hormone, beta Subunit/genetics , Gene Expression Regulation/physiology , Inhibin-beta Subunits/physiology , Transcription, Genetic/physiology , Animals , Base Sequence , Cell Line , DNA , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Sequence Homology, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL