Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 346
Filter
1.
Biochem Pharmacol ; 214: 115668, 2023 08.
Article in English | MEDLINE | ID: mdl-37364623

ABSTRACT

Activins are a subgroup of the TGFß superfamily of growth and differentiation factors, dimeric in nature and consisting of two inhibin beta subunits linked via a disulfide bridge. Canonical activin signaling occurs through Smad2/3, with negative feedback initiated by Smad6/7 following signal transduction, which binds activin type I receptor preventing phosphorylation of Smad2/3 and activation of downstream signaling. In addition to Smad6/7, other inhibitors of activin signaling have been identified as well, including inhibins (dimers of an inhibin alpha and beta subunit), BAMBI, Cripto, follistatin, and follistatin-like 3 (fstl3). To date, activins A, B, AB, C, and E have been identified and isolated in mammals, with activin A and B having the most characterization of biological activity. Activin A has been implicated as a regulator of several important functions of liver biology, including hepatocyte proliferation and apoptosis, ECM production, and liver regeneration; the role of other subunits of activin in liver physiology are less understood. There is mounting data to suggest a link between dysregulation of activins contributing to various hepatic diseases such as inflammation, fibrosis, and hepatocellular carcinoma, and emerging studies demonstrating the protective and regenerative effects of inhibiting activins in mouse models of liver disease. Due to their importance in liver biology, activins demonstrate utility as a therapeutic target for the treatment of hepatic diseases such as cirrhosis, NASH, NAFLD, and HCC; further research regarding activins may provide diagnostic or therapeutic opportunity for those suffering from various liver diseases.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Mice , Animals , Follistatin , Activins/physiology , Activin Receptors , Mammals
2.
Front Immunol ; 11: 2115, 2020.
Article in English | MEDLINE | ID: mdl-33117332

ABSTRACT

The epididymis is an important male accessory sex organ where sperm motility and fertilization ability develop. When spermatozoa carrying foreign antigens enter the epididymis, the epididymis shows "immune privilege" to tolerate them. It is well-known that a tolerogenic environment exists in the caput epididymis, while pro-inflammatory circumstances prefer the cauda epididymis. This meticulously regulated immune environment not only protects spermatozoa from autoimmunity but also defends spermatozoa against pathogenic damage. Epididymitis is one of the common causes of male infertility. Up to 40% of patients suffer from permanent oligospermia or azoospermia. This is related to the immune characteristics of the epididymis itself. Moreover, epididymitis induced by different pathogenic microbial infections has different characteristics. This article elaborates on the distribution and immune response characteristics of epididymis immune cells, the role of epididymis epithelial cells (EECs), and the epididymis defense against different pathogenic infections (such as uropathogenic Escherichia coli, Chlamydia trachomatis, and viruses to provide therapeutic approaches for epididymitis and its subsequent fertility problems.


Subject(s)
Epididymis/immunology , Epididymitis/immunology , Spermatozoa/immunology , Activins/physiology , Acute Disease , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Blood-Testis Barrier , Chlamydia Infections/immunology , Chlamydia trachomatis/immunology , Defensins/physiology , Epididymitis/complications , Epididymitis/epidemiology , Epididymitis/microbiology , Escherichia coli Infections/immunology , Humans , Immune System/cytology , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Infertility, Male/etiology , Infertility, Male/immunology , Infertility, Male/microbiology , Male , Mice , Middle Aged , TGF-beta Superfamily Proteins/physiology , Uropathogenic Escherichia coli/immunology , Virus Diseases/immunology , Young Adult
3.
Int J Mol Sci ; 21(18)2020 Sep 05.
Article in English | MEDLINE | ID: mdl-32899497

ABSTRACT

Activins transduce the TGF-ß pathway through a heteromeric signaling complex consisting of type I and type II receptors, and activins also inhibit bone morphogenetic protein (BMP) signaling mediated by type I receptor ALK2. Recent studies indicated that activin A cross-activates the BMP pathway through ALK2R206H, a mutation associated with Fibrodysplasia Ossificans Progressiva (FOP). How activin A inhibits ALK2WT-mediated BMP signaling but activates ALK2R206H-mediated BMP signaling is not well understood, and here we offer some insights into its molecular mechanism. We first demonstrated that among four BMP type I receptors, ALK2 is the only subtype able to mediate the activin A-induced BMP signaling upon the dissociation of FKBP12. We further showed that BMP4 does not cross-signal TGF-ß pathway upon FKBP12 inhibition. In addition, although the roles of type II receptors in the ligand-independent BMP signaling activated by FOP-associated mutant ALK2 have been reported, their roles in activin A-induced BMP signaling remains unclear. We demonstrated in this study that the known type II BMP receptors contribute to activin A-induced BMP signaling through their kinase activity. Together, the current study provided important mechanistic insights at the molecular level into further understanding physiological and pathophysiological BMP signaling.


Subject(s)
Activin Receptors, Type I/metabolism , Activins/metabolism , Bone Morphogenetic Proteins/metabolism , Activin Receptors, Type I/genetics , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Activins/physiology , Animals , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Proteins/physiology , Cell Differentiation/physiology , Cell Line , Gene Expression Regulation/genetics , HEK293 Cells , Hep G2 Cells , Humans , Mice , Ossification, Heterotopic/genetics , Phosphorylation , Signal Transduction/genetics , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism
4.
Mol Cell Endocrinol ; 517: 110963, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32745576

ABSTRACT

Anti-Müllerian hormone (AMH/Amh) plays a role in gonadal differentiation and function across vertebrates. In zebrafish we demonstrated that Amh deficiency caused severe gonadal dysgenesis and dysfunction. The mutant gonads showed extreme hypertrophy with accumulation of early germ cells in both sexes, namely spermatogonia in the testis and primary growth oocytes in the ovary. In amh mutant females, the folliculogenesis was normal in young fish but receded progressively in adults, which was accompanied by progressive decrease in follicle-stimulating hormone (fshb) expression. Interestingly the expression of fshb increased in the pituitary of juvenile amh mutant males but decreased in adults. The upregulation of fshb in mutant male juveniles was likely one of the mechanisms for triggering gonadal hypergrowth, whereas the downregulation of fshb in adults might involve a negative feedback by gonadal inhibin. Further analysis using mutants of fshb and growth differentiation factor 9 (gdf9) provided evidence for a role of FSH in triggering ovarian hypertrophy in young female amh mutant as well. In summary, the present study provided comprehensive genetic evidence for dual roles of Amh in controlling zebrafish gonadal homeostasis and gametogenesis in both sexes. Amh suppresses proliferation or accumulation of early germ cells (spermatogonia in testis and primary growth oocytes in ovary) while promoting their exit to advanced stages, and its action may involve both endocrine and paracrine pathways.


Subject(s)
Anti-Mullerian Hormone/physiology , Gametogenesis/physiology , Homeostasis/physiology , Zebrafish Proteins/physiology , Activins/physiology , Animals , Anti-Mullerian Hormone/deficiency , Anti-Mullerian Hormone/genetics , Base Sequence , CRISPR-Cas Systems , Feedback, Physiological , Female , Follicle Stimulating Hormone/biosynthesis , Follicle Stimulating Hormone/genetics , Gene Knockout Techniques , Growth Differentiation Factor 9/genetics , Hypertrophy , Infertility, Female/genetics , Infertility, Male/genetics , Inhibins/physiology , Male , Ovary/metabolism , Ovary/pathology , Paracrine Communication , Pituitary Gland, Anterior/metabolism , Sexual Maturation/genetics , Testis/metabolism , Testis/pathology , Zebrafish
5.
Endocrinology ; 161(7)2020 07 01.
Article in English | MEDLINE | ID: mdl-32274496

ABSTRACT

Activin A promotes fetal mouse testis development, including driving Sertoli cell proliferation and cord morphogenesis, but its mechanisms of action are undefined. We performed ribonucleic acid sequencing (RNA-seq) on testicular somatic cells from fetal activin A-deficient mice (Inhba KO) and wildtype littermates at embryonic day (E) E13.5 and E15.5. Analysis of whole gonads provided validation, and cultures with a pathway inhibitor discerned acute from chronic effects of altered activin A bioactivity. Activin A deficiency predominantly affects the Sertoli cell transcriptome. New candidate targets include Minar1, Sel1l3, Vnn1, Sfrp4, Masp1, Nell1, Tthy1 and Prss12. Importantly, the testosterone (T) biosynthetic enzymes present in fetal Sertoli cells, Hsd17b1 and Hsd17b3, were identified as activin-responsive. Activin-deficient testes contained elevated androstenedione (A4), displayed an Inhba gene dose-dependent A4/T ratio, and contained 11-keto androgens. The remarkable accumulation of lipid droplets in both Sertoli and germ cells at E15.5 indicated impaired lipid metabolism in the absence of activin A. This demonstrated for the first time that activin A acts on Sertoli cells to determine local steroid production during fetal testis development. These outcomes reveal how compounds that perturb fetal steroidogenesis can function through cell-specific mechanisms and can indicate how altered activin levels in utero may impact testis development.


Subject(s)
Activins/physiology , Gonadal Steroid Hormones/metabolism , Testis/embryology , Testis/metabolism , Animals , Female , Male , Mice , Mice, Knockout , Pregnancy , Sex Determination Processes
6.
Curr Opin Nephrol Hypertens ; 29(1): 136-144, 2020 01.
Article in English | MEDLINE | ID: mdl-31714286

ABSTRACT

PURPOSE OF REVIEW: This review highlights recent discoveries and advances that have been made in understanding the role of the TGFß superfamily members activins, and in particular, activin A (ActA), in renal disease. RECENT FINDINGS: A deleterious role for ActA in renal disease and its complications has begun to emerge. We summarize data supporting an important contribution of ActA to kidney fibrosis and inflammation of varying causes, as well as its role in the development of a particular bone mineral disorder seen in chronic kidney disease (CKD) called mineral bone disorder (MBD), including vascular calcification. Finally, we discuss ActA in the context of anemia associated with chronic kidney disease and review potential approaches to treatment based on ActA blockade. SUMMARY: ActA is an important contributor to the pathogenesis of acute and chronic kidney disease of varying causes. Preclinical studies show that ActA inhibition, through various approaches, is protective in rodent models of kidney disease. The potential adverse effects of some of these approaches can be attributed to their targeting of other TGFß family ligands. Further preclinical and clinical investigations testing the therapeutic efficacy of more selective ActA inhibition on the progression of acute and chronic kidney disease and its impact on bone-mineral disorder would more definitively establish its role in renal disease.


Subject(s)
Activins/physiology , Kidney Diseases/etiology , Chronic Kidney Disease-Mineral and Bone Disorder/etiology , Humans , Vascular Calcification/etiology
7.
Sci Rep ; 9(1): 17359, 2019 11 22.
Article in English | MEDLINE | ID: mdl-31757999

ABSTRACT

Tumor angiogenesis is a key factor in the progression of thymic epithelial tumors (TETs). Activin A, a member of the TGFß family, and its antagonist Follistatin are involved in several human malignancies and angiogenesis. We investigated Activin A and Follistatin in serum and tumor tissue of patients with TETs in relation to microvessel density (MVD), WHO histology classification, tumor stage and outcome. Membranous Activin A expression was detected in all tumor tissues of TETs, while Follistatin staining was found in tumor nuclei and cytoplasm. Patients with TETs presented with significantly higher Activin A and Follistatin serum concentrations compared to healthy volunteers, respectively. Follistatin serum concentrations correlated significantly with tumor stage and decreased to physiologic values after complete tumor resection. Follistatin serum concentrations correlated further with MVD and were associated with significantly worse freedom from recurrence (FFR). Low numbers of immature tumor vessels represented even an independent worse prognostic factor for FFR at multivariable analysis. To conclude, the Activin A - Follistatin axis is involved in the pathogenesis of TETs. Further study of Follistatin and Activin A in TETs is warranted as the molecules may serve as targets to inhibit tumor angiogenesis and tumor progression.


Subject(s)
Activins/physiology , Follistatin/physiology , Neoplasms, Glandular and Epithelial/diagnosis , Neovascularization, Pathologic/etiology , Thymus Gland/blood supply , Thymus Neoplasms/diagnosis , Activins/blood , Activins/metabolism , Adult , Aged , Case-Control Studies , Disease Progression , Female , Follistatin/blood , Follistatin/metabolism , Humans , Male , Middle Aged , Myasthenia Gravis/blood , Myasthenia Gravis/metabolism , Myasthenia Gravis/surgery , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Neoplasms, Glandular and Epithelial/surgery , Neovascularization, Pathologic/diagnosis , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Postoperative Period , Prognosis , Thymus Gland/abnormalities , Thymus Gland/metabolism , Thymus Gland/pathology , Thymus Gland/surgery , Thymus Neoplasms/metabolism , Thymus Neoplasms/pathology , Thymus Neoplasms/surgery
8.
Geroscience ; 41(3): 267-273, 2019 06.
Article in English | MEDLINE | ID: mdl-31243699

ABSTRACT

Women live longer than men in virtually all circumstances. However, a more common pattern among animals is that one sex lives longer under some conditions, the other lives longer under other conditions. In laboratory mice, interventions that extend longevity are surprisingly often sex-specific in their effects. Understanding these conditional sex differences could provide mechanistic insight into how longevity could be modulated in humans. One way that longevity can be consistently enhanced is by inhibiting reproduction or eliminating the capacity to reproduce. Thus, there appears to be a mechanistic link between gonadal activity and longevity. There also appears to be a mechanistic link between some types of neuroendocrine signaling and longevity. Combining these two observations suggest that communication between the brain and gonad is a ripe avenue for further exploring longevity-assurance mechanisms. Also, because the timing and activity of specific brain-gonad endocrine differs between the sexes, neuroendocrine linkages between the brain and gonad, particularly among the less obvious hormones such as activin and inhibin, could provide additional insight into mechanisms of sex differences in aging.


Subject(s)
Aging/physiology , Brain/physiopathology , Gonads/physiopathology , Sex Characteristics , Activins/physiology , Animals , Female , Humans , Hypothalamo-Hypophyseal System/physiology , Inhibins/physiology , Longevity/physiology , Male , Neurosecretory Systems/physiology , Reproduction/physiology
9.
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
10.
J Biol Chem ; 293(48): 18444-18453, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30282636

ABSTRACT

Activin and Wnt signaling are necessary and sufficient for mesendoderm (ME) differentiation of human embryonic stem cells (ESCs). In this study, we report that during ME differentiation induced by Activin and Wnt, Activin/Smad2 induces a decrease of the repressive histone modification of H3K27me3 by promoting the proteasome-dependent degradation of enhancer of zeste 2 polycomb (EZH2)-repressive complex 2 subunit. As a result, recruitment of the forkhead protein FOXH1 on open chromatin regions integrates the signals of Activin/Smad2 and Wnt/ß-catenin to activate the expression of the ME genes including HAS2 and ALDH3A2 Consistently, H3K27me3 decrease is enriched on open chromatin around regulatory regions. Furthermore, knockdown of HAS2 or ALDH3A2 greatly attenuates ME differentiation. These findings unveil a pathway from extracellular signals to epigenetic modification-mediated gene activation during ME commitment.


Subject(s)
Activins/physiology , Aldehyde Oxidoreductases/physiology , Cell Differentiation/physiology , Endoderm/cytology , Human Embryonic Stem Cells/cytology , Hyaluronan Synthases/physiology , Mesoderm/cytology , Smad2 Protein/physiology , Up-Regulation , Wnt Signaling Pathway , beta Catenin/physiology , Chromatin/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic , Forkhead Transcription Factors/metabolism , Histones/metabolism , Humans , Promoter Regions, Genetic , Proteolysis
11.
J Clin Endocrinol Metab ; 103(10): 3890-3899, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30085147

ABSTRACT

Context: Clinical trials are evaluating the efficacy of inhibitors of the myostatin pathway in neuromuscular and metabolic diseases. Activins and follistatins are major regulators of the myostatin pathway, but their physiology in relation to metabolic and anthropometric variables and in response to exercise remains to be fully elucidated in humans. Objective: We investigated whether concentrations of circulating activin A, activin B, follistatin, and follistatin-like 3 (FSTL3) are associated with anthropometric and metabolic variables and whether they are affected by exercise. Design: Activin A, activin B, follistatin, and FSTL3 were measured in (1) 80 subjects divided according to age (young vs old) and fitness status (active vs sedentary) before and after exercise at 70% maximal oxygen consumption (VO2max), followed by 90% of VO2max until exhaustion; and (2) 23 subjects [9 healthy and 14 with metabolic syndrome (MetS)] who completed four sessions: no exercise, high-intensity interval exercise, continuous moderate-intensity exercise, and resistance exercise for up to 45 minutes. Results: At baseline, follistatin and FSTL3 concentrations were positively associated with age, fat percentage, and body mass index (P < 0.001). Follistatin was positively associated with serum cholesterol (P = 0.005), low-density lipoprotein cholesterol (P = 0.01), triglycerides (P = 0.033), and blood pressure (P = 0.019), whereas activin A and activin B were higher in physically active participants (P = 0.056 and 0.029, respectively). All exercise types increased the levels of all hormones ∼10% to 21% (P = 0.034 for activin B, P < 0.001 for the others) independent of the presence of MetS. Conclusion: Concentrations of circulating activins and follistatins are associated with metabolic parameters and increase after 45 minutes of exercise.


Subject(s)
Activins/physiology , Exercise/physiology , Follistatin/physiology , Activins/blood , Adiposity/physiology , Adolescent , Adult , Aged , Aging/blood , Aging/physiology , Anthropometry/methods , Body Composition/physiology , Follistatin/blood , Follistatin-Related Proteins/blood , Follistatin-Related Proteins/physiology , Humans , Male , Middle Aged , Physical Fitness/physiology , Sedentary Behavior , Young Adult
12.
Calcif Tissue Int ; 102(2): 251-264, 2018 02.
Article in English | MEDLINE | ID: mdl-29353450

ABSTRACT

Considerable advances in oncology over recent decades have led to improved survival, while raising concerns about long-term consequences of anticancer treatments. In patients with breast or prostate malignancies, bone health is a major issue due to the high risk of bone metastases and the frequent prolonged use of hormone therapies that alter physiological bone turnover, leading to increased fracture risk. Thus, the onset of cancer treatment-induced bone loss (CTIBL) should be considered by clinicians and recent guidelines should be routinely applied to these patients. In particular, baseline and periodic follow-up evaluations of bone health parameters enable the identification of patients at high risk of osteoporosis and fractures, which can be prevented by the use of bone-targeting agents (BTAs), calcium and vitamin D supplementation and modifications of lifestyle. This review will focus upon the pathophysiology of breast and prostate cancer treatment-induced bone loss and the most recent evidence about effective preventive and therapeutic strategies.


Subject(s)
Antineoplastic Agents/adverse effects , Bone and Bones/drug effects , Breast Neoplasms/complications , Osteoporosis/chemically induced , Prostatic Neoplasms/complications , Activins/physiology , Androgens/physiology , Antineoplastic Agents/therapeutic use , Bone and Bones/physiology , Bone and Bones/physiopathology , Breast Neoplasms/drug therapy , Breast Neoplasms/physiopathology , Estrogens/physiology , Female , Gonadotropin-Releasing Hormone/agonists , Humans , Inhibins/physiology , Male , Osteoporosis/physiopathology , Practice Guidelines as Topic , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/physiopathology
13.
Biochim Biophys Acta Mol Basis Dis ; 1864(3): 891-899, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29287776

ABSTRACT

BACKGROUND & AIMS: TGFß superfamily member Activin-A is a multifunctional hormone/cytokine expressed in multiple tissues and cells, where it regulates cellular differentiation, proliferation, inflammation and tissue architecture. High activin-A levels have been reported in alcoholic cirrhosis and non-alcoholic steatohepatitis (NASH). Our aim was to identify the cell types involved in the fibrotic processes induced by activin-A in liver and verify the liver diseases that this molecule can be found increased. METHODS: We studied the effect of activin-A on mouse primary Kupffer cells (KCs) and Hepatic Stellate cells (HSCs) and the levels of activin-A and its inhibitor follistatin in the serum of patients from a large panel of liver diseases. RESULTS: Activin-A is expressed by mouse hepatocytes, HSCs and Liver Sinusoid Endothelial cells but not KCs. Each cell type expresses different activin receptor combinations. HSCs are unresponsive to activin-A due to downregulation/desensitization of type-II activin receptors, while KCs respond by increasing the expression/production of TNFα και TGFß1. In the presence of KCs or conditioned medium from activin-A treated KCs, HSCs switch to a profibrogenic phenotype, including increased collagen and αSMA expression and migratory capacity. Incubation of activin-A treated KC conditioned medium with antibodies against TNFα and TGFß1 partially blocks its capacity to activate HSCs. Only patients with alcoholic liver diseases and NASH cirrhosis have significantly higher activin-A levels and activin-A/follistatin ratio. CONCLUSIONS: Activin-A may induce fibrosis in NASH and alcoholic cirrhosis via activation of KCs to express pro-inflammatory molecules that promote HSC-dependent fibrogenesis and could be a target for future anti-fibrotic therapies.


Subject(s)
Activins/physiology , Hepatic Stellate Cells/metabolism , Kupffer Cells/metabolism , Liver/pathology , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Activins/genetics , Activins/metabolism , Aged , Animals , Case-Control Studies , Fibrosis/genetics , Fibrosis/metabolism , Humans , Kupffer Cells/pathology , Liver/metabolism , Liver Cirrhosis/metabolism , Male , Mice , Mice, Inbred BALB C , Middle Aged , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Transforming Growth Factor beta/genetics , Tumor Necrosis Factor-alpha/genetics
14.
Genes Dev ; 31(22): 2250-2263, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29269485

ABSTRACT

Activin/SMAD signaling in human embryonic stem cells (hESCs) ensures NANOG expression and stem cell pluripotency. In the presence of Wnt ligand, the Activin/SMAD transcription network switches to cooperate with Wnt/ß-catenin and induce mesendodermal (ME) differentiation genes. We show here that the Hippo effector YAP binds to the WNT3 gene enhancer and prevents the gene from being induced by Activin in proliferating hESCs. ChIP-seq (chromatin immunoprecipitation [ChIP] combined with high-throughput sequencing) data show that YAP impairs SMAD recruitment and the accumulation of P-TEFb-associated RNA polymerase II (RNAPII) C-terminal domain (CTD)-Ser7 phosphorylation at the WNT3 gene. CRISPR/CAS9 knockout of YAP in hESCs enables Activin to induce Wnt3 expression and stabilize ß-catenin, which then synergizes with Activin-induced SMADs to activate a subset of ME genes that is required to form cardiac mesoderm. Interestingly, exposure of YAP-/- hESCs to Activin induces cardiac mesoderm markers (BAF60c and HAND1) without activating Wnt-dependent cardiac inhibitor genes (CDX2 and MSX1). Moreover, canonical Wnt target genes are up-regulated only modestly, if at all, under these conditions. Consequently, YAP-null hESCs exposed to Activin differentiate precisely into beating cardiomyocytes without further treatment. We conclude that YAP maintains hESC pluripotency by preventing WNT3 expression in response to Activin, thereby blocking a direct route to embryonic cardiac mesoderm formation.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Myocytes, Cardiac/metabolism , Nuclear Proteins/physiology , Repressor Proteins/physiology , Transcription Factors/physiology , Wnt3 Protein/genetics , Activins/physiology , CDX2 Transcription Factor/genetics , Cell Cycle Proteins , Cell Differentiation/genetics , Cell Lineage , Cells, Cultured , Chromatin/metabolism , Embryonic Stem Cells/cytology , Enhancer Elements, Genetic , Heart/embryology , Humans , Mesoderm/cytology , Nuclear Proteins/genetics , Promoter Regions, Genetic , Repressor Proteins/genetics , Signal Transduction , Smad Proteins/antagonists & inhibitors , Transcription Elongation, Genetic , Transcription Factors/genetics , beta Catenin/metabolism
15.
J Dent Res ; 96(10): 1145-1152, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28605600

ABSTRACT

Previous studies show that both activin and Bmp4 act as crucial mesenchymal odontogenic signals during early tooth development. Remarkably, mice lacking activin-ßA ( Inhba-/-) and mice with neural crest-specific inactivation of Bmp4 ( Bmp4ncko/ncko) both exhibit bud-stage developmental arrest of the mandibular molar tooth germs while their maxillary molar tooth germs completed morphogenesis. In this study, we found that, whereas expression of Inhba and Bmp4 in the developing tooth mesenchyme is independent of each other, Bmp4ncko/nckoInhba-/- compound mutant mice exhibit early developmental arrest of all tooth germs. Moreover, genetic inactivation of Osr2, a negative regulator of the odontogenic function of the Bmp4-Msx1 signaling pathway, rescues mandibular molar morphogenesis in Inhba-/- embryos. We recently reported that Osr2 and the Bmp4-Msx1 pathway control the bud-to-cap transition of tooth morphogenesis through antagonistic regulation of expression of secreted Wnt antagonists, including Dkk2 and Sfrp2, in the developing tooth mesenchyme. We show here that expression of Dkk2 messenger RNAs was significantly upregulated and expanded into the tooth bud mesenchyme in Inhba-/- embryos in comparison with wild-type littermates. Furthermore, in utero treatment with either lithium chloride, an agonist of canonical Wnt signaling, or the DKK inhibitor IIIC3a rescued mandibular molar tooth morphogenesis in Inhba-/- embryos. Together with our finding that the developing mandibular molar tooth bud mesenchyme expresses significantly higher levels of Dkk2 than the developing maxillary molar tooth mesenchyme, these data indicate that Bmp4 and activin signaling pathways converge on activation of the Wnt signaling pathway to promote tooth morphogenesis through the bud-to-cap transition and that the differential effects of loss of activin or Bmp4 signaling on maxillary and mandibular molar tooth morphogenesis are mainly due to the differential expression of Wnt antagonists, particularly Dkk2, in the maxillary and mandibular tooth mesenchyme.


Subject(s)
Activins/physiology , Bone Morphogenetic Protein 4/physiology , Molar/embryology , Odontogenesis/physiology , Tooth Germ/embryology , Wnt Signaling Pathway/physiology , Animals , Gene Expression Regulation, Developmental , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/metabolism , Laser Capture Microdissection , Lithium Chloride/pharmacology , Membrane Proteins/metabolism , Mice , Mice, Transgenic , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction , Transcription Factors/metabolism , Transcription Factors/physiology
16.
PLoS One ; 12(6): e0179789, 2017.
Article in English | MEDLINE | ID: mdl-28662143

ABSTRACT

Gonadotrophin regulation by activin/follistatin system is well-documented, but the corresponding effect on growth hormone (GH) has not been fully characterized and with little information available in lower vertebrates, especially in fish models. In grass carp, local interactions of GH and luteinizing hormone (LH) can induce GH release and gene expression at pituitary level via autocrine/paracrine mechanisms. To shed light on the role of activin/follistatin system in GH regulation by local actions of GH and LH, grass carp activin ßA and ßB were cloned, shown to be single-copy genes expressed in the pituitary, and confirmed to encode activin proteins capable of transactivating promoter with activin-responsive elements. In grass carp pituitary cells, activin A and B were effective in reducing GH secretion and GH cell content with concurrent drop in GH mRNA level whereas the opposite was true for follistatin, the activin-binding protein known to neutralize the effects of endogenous activin. Treatment with activin A and B not only could suppress basal but also inhibit GH mRNA expression induced by GH and human chorionic gonadotropin (hCG), a functional analogue of LH in fish model. Apparently, down-regulation of GH mRNA by activin was mediated by reducing GH transcript stability with concurrent inhibition on GH promoter activity via the SMAD pathway. In reciprocal experiments, GH treatment was found to up-regulate activin ßA, activin ßB and follistatin mRNA levels in carp pituitary cells but the opposite was noted by removing endogenous GH with GH antiserum. Interestingly, parallel treatment with hCG could also inhibit basal as well as GH-induced activin ßA, activin ßB and follistatin gene expression. These results, as a whole, indicate that the pituitary activin/follistatin system can serve as a regulatory target for local interactions of GH and LH and contribute to GH regulation by autocrine/paracrine mechanisms in the carp pituitary.


Subject(s)
Activins/physiology , Follistatin/physiology , Growth Hormone/metabolism , Luteinizing Hormone/metabolism , Pituitary Gland/physiology , Activins/genetics , Animals , Carps , Cloning, Molecular , Female , Follistatin/genetics , Male , Pituitary Gland/cytology
17.
Reprod Domest Anim ; 52 Suppl 2: 29-34, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28111799

ABSTRACT

Canid reproduction is unique among other mammals in that females experience long and variable periods of ovarian inactivity. While the domestic dog exhibits a non-seasonal, largely sporadic monoestrus occurring once or twice a year, most wild canids, such as the gray wolf (Canis lupus) and red wolf (Canis rufus), are seasonal breeders with onset apparently dependent on species, latitudinal location and/or variety of environment factors. Neuroendocrine controls of ovarian functions have been mostly studied in the dog, but less so in their wild counterparts, due to difficulties in regular blood sampling. Yet, development of non-invasive hormone monitoring has advanced the understanding of reproductive cycle in wild canids. Recent advances in in vitro follicle culture technology also have begun to provide insights into paracrine controls of canid ovarian folliculogenesis. This review highlights current knowledge on canid reproduction with emphasis on endocrine and paracrine controls of follicular development. We also discuss future research priorities, including advancing the understanding of anoestrous termination and role of paracrine factors in canine folliculogenesis.


Subject(s)
Canidae/physiology , Follicle Stimulating Hormone/physiology , Luteinizing Hormone/physiology , Ovarian Follicle/physiology , Reproduction/physiology , Activins/physiology , Animals , Female , Growth Hormone/physiology , Insulin/physiology , Insulin-Like Growth Factor I/physiology
18.
Reprod Fertil Dev ; 29(7): 1329-1339, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27185102

ABSTRACT

The reproductive tract secretes bioactive molecules collectively known as embryokines that can regulate embryonic growth and development. In the present study we tested four growth factors expressed in the endometrium for their ability to modify the development of the bovine embryo to the blastocyst stage and alter the expression of genes found to be upregulated (bone morphogenetic protein 15 (BMP15) and keratin 8, type II (KRT8)) or downregulated (NADH dehydrogenase 1 (ND1) and S100 calcium binding protein A10 (S100A10)) in embryos competent to develop to term. Zygotes were treated at Day 5 with 0.01, 0.1 or 1.0nM growth factor. The highest concentration of activin A increased the percentage of putative zygotes that developed to the blastocyst stage. Connective tissue growth factor (CTGF) increased the number of cells in the inner cell mass (ICM), decreased the trophectoderm:ICM ratio and increased blastocyst expression of KRT8 and ND1. The lowest concentration of hepatocyte growth factor (HGF) reduced the percentage of putative zygotes becoming blastocysts. Teratocarcinoma-derived growth factor 1 increased total cell number at 0.01nM and expression of S100A10 at 1.0nM, but otherwise had no effects. Results confirm the prodevelopmental actions of activin A and indicate that CTGF may also function as an embryokine by regulating the number of ICM cells in the blastocyst and altering gene expression. Low concentrations of HGF were inhibitory to development.


Subject(s)
Activins/physiology , Blastocyst/physiology , Cattle/embryology , Cattle/physiology , Connective Tissue Growth Factor/physiology , Embryonic Development/physiology , Hepatocyte Growth Factor/physiology , Activins/pharmacology , Animals , Blastocyst/drug effects , Connective Tissue Growth Factor/pharmacology , Embryo Culture Techniques/methods , Embryo Culture Techniques/veterinary , Embryonic Development/drug effects , Female , Growth Substances/pharmacology , Growth Substances/physiology , Hepatocyte Growth Factor/pharmacology , Pregnancy
19.
Dev Biol ; 426(2): 429-441, 2017 06 15.
Article in English | MEDLINE | ID: mdl-27209239

ABSTRACT

During early vertebrate embryogenesis, cell fate specification is often coupled with cell acquisition of specific adhesive, polar and/or motile behaviors. In Xenopus gastrulae, tissues fated to form different axial structures display distinct motility. The cells in the early organizer move collectively and directionally toward the animal pole and contribute to anterior mesendoderm, whereas the dorsal and the ventral-posterior trunk tissues surrounding the blastopore of mid-gastrula embryos undergo convergent extension and convergent thickening movements, respectively. While factors regulating cell lineage specification have been described in some detail, the molecular machinery that controls cell motility is not understood in depth. To gain insight into the gene battery that regulates both cell fates and motility in particular embryonic tissues, we performed RNA sequencing (RNA-seq) to investigate differentially expressed genes in the early organizer, the dorsal and the ventral marginal zone of Xenopus gastrulae. We uncovered many known signaling and transcription factors that have been reported to play roles in embryonic patterning during gastrulation. We also identified many uncharacterized genes as well as genes that encoded extracellular matrix (ECM) proteins or potential regulators of actin cytoskeleton. Co-expression of a selected subset of the differentially expressed genes with activin in animal caps revealed that they had distinct ability to block activin-induced animal cap elongation. Most of these factors did not interfere with mesodermal induction by activin, but an ECM protein, EFEMP2, inhibited activin signaling and acted downstream of the activated type I receptor. By focusing on a secreted protein kinase PKDCC1, we showed with overexpression and knockdown experiments that PKDCC1 regulated gastrulation movements as well as anterior neural patterning during early Xenopus development. Overall, our studies identify many differentially expressed signaling and cytoskeleton regulators in different embryonic regions of Xenopus gastrulae and imply their functions in regulating cell fates and/or behaviors during gastrulation.


Subject(s)
Body Patterning/genetics , Gastrula/metabolism , Gene Expression Regulation, Developmental/genetics , Sequence Analysis, RNA , Xenopus/genetics , Activins/physiology , Animals , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/ultrastructure , Extracellular Matrix Proteins/physiology , Gastrula/ultrastructure , Germ Layers/metabolism , Morphogenesis/genetics , Organizers, Embryonic , Protein-Tyrosine Kinases/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Xenopus/embryology , Xenopus Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...