128 results on '"Activins antagonists & inhibitors"'
Search Results
2. Sotatercept: First Approval.
- Author
-
Kang C
- Subjects
- Humans, Recombinant Fusion Proteins therapeutic use, Recombinant Fusion Proteins pharmacology, United States, Activins antagonists & inhibitors, Hypertension, Pulmonary drug therapy, Pulmonary Arterial Hypertension drug therapy, Drug Approval
- Abstract
Sotatercept (sotatercept-csrk; WINREVAIR
TM ) is an activin signalling inhibitor that is being developed by Merck and Co., Inc. (Rahway, NJ, USA) for the treatment of pulmonary arterial hypertension. Sotatercept recently received approval in the USA for the treatment of adults with pulmonary arterial hypertension [World Health Organisation (WHO) Group 1] to increase exercise capacity, improve WHO functional class and reduce the risk of clinical worsening events. This article summarizes the milestones in the development of sotatercept leading to this first approval for pulmonary arterial hypertension., (© 2024. The Author(s), under exclusive licence to Springer Nature Switzerland AG.)- Published
- 2024
- Full Text
- View/download PDF
3. Activin A and Cell-Surface GRP78 Are Novel Targetable RhoA Activators for Diabetic Kidney Disease.
- Author
-
Soomro A, Trink J, O'Neil K, Li R, Naiel S, Gao B, Ask K, and Krepinsky JC
- Subjects
- Activins antagonists & inhibitors, Activins metabolism, Animals, Antibodies, Neutralizing pharmacology, Diabetes Mellitus, Experimental chemically induced, Diabetes Mellitus, Experimental drug therapy, Diabetes Mellitus, Experimental metabolism, Diabetic Nephropathies drug therapy, Diabetic Nephropathies genetics, Diabetic Nephropathies metabolism, Diabetic Nephropathies pathology, Disease Models, Animal, Endoplasmic Reticulum Chaperone BiP, Follistatin pharmacology, Gene Expression Regulation, Heat-Shock Proteins antagonists & inhibitors, Heat-Shock Proteins metabolism, Humans, Male, Mesangial Cells drug effects, Mesangial Cells pathology, Mice, Mice, Inbred C57BL, Nephrectomy methods, Primary Cell Culture, Signal Transduction, Streptozocin administration & dosage, rho-Associated Kinases genetics, rho-Associated Kinases metabolism, rhoA GTP-Binding Protein metabolism, Activins genetics, Diabetes Mellitus, Experimental genetics, Heat-Shock Proteins genetics, Mesangial Cells metabolism, rhoA GTP-Binding Protein genetics
- Abstract
Diabetic kidney disease (DKD) is the leading cause of kidney failure. RhoA/Rho-associated protein kinase (ROCK) signaling is a recognized mediator of its pathogenesis, largely through mediating the profibrotic response. While RhoA activation is not feasible due to the central role it plays in normal physiology, ROCK inhibition has been found to be effective in attenuating DKD in preclinical models. However, this has not been evaluated in clinical studies as of yet. Alternate means of inhibiting RhoA/ROCK signaling involve the identification of disease-specific activators. This report presents evidence showing the activation of RhoA/ROCK signaling both in vitro in glomerular mesangial cells and in vivo in diabetic kidneys by two recently described novel pathogenic mediators of fibrosis in DKD, activins and cell-surface GRP78. Neither are present in normal kidneys. Activin inhibition with follistatin and neutralization of cell-surface GRP78 using a specific antibody blocked RhoA activation in mesangial cells and in diabetic kidneys. These data identify two novel RhoA/ROCK activators in diabetic kidneys that can be evaluated for their efficacy in inhibiting the progression of DKD.
- Published
- 2021
- Full Text
- View/download PDF
4. Inhibition of Activin A suppressed tumor necrosis factor-α secretion and improved histopathological conditions in malarial mice.
- Author
-
Chin VK, Tie TH, Abd Majid R, Hassan H, Nordin N, Abas R, and Basir R
- Subjects
- Activins immunology, Animals, Cytokines blood, Interferon-gamma blood, Interleukin-10 blood, Malaria immunology, Male, Mice, Mice, Inbred ICR, Plasmodium berghei, Activins antagonists & inhibitors, Malaria drug therapy, Tumor Necrosis Factor-alpha blood
- Abstract
Malaria infection still remains as one of the most prominent parasitic diseases afflicting mankind in tropical and subtropical regions. The severity of malaria infection has often been associated to exuberant host immune inflammatory responses that could possibly lead to severe immunopathological conditions and subsequent death of host tissues. Activin A is a protein belonging to the transforming growth factor-beta (TGF-β) family that regulates multiple physiological processes and pathological-associated diseases. The biological roles of activin A have been associated with manipulation of inflammation-related processes and modulation of host immune responses. This implies that activin A protein could play a role in malaria pathogenesis since malaria infection has been closely linked to severe immune responses leading to death, However, the actual in vivo role of activin A in malaria infection remains elusive. Hence, this study was undertaken to investigate the involvement of activin A in malaria infection as well as to assess the modulating effects of activin A on the cytokine releases (TNF-α, IFN-γ and IL-10) and histopathological changes in major affected organs (kidney, liver, lung, brain and spleen) in malarial mice infected with Plasmodium berghei ANKA. Our results showed that the concentrations of plasma activin A were significantly increased in malarial mice throughout the study periods. Also. the systemic activin A level was positively correlated with malaria parasitemia. This indicates that activin A could play a role in malaria pathogenesis and malaria parasitemia development. Plasma TNF-α, IFN-γ and IL-10 cytokine levels were significantly increased in malarial mice at day-5 post infection, suggesting that these cytokines attributed to severe malaria pathogenesis. Histopathological features such as sequestration of parasitized red blood cells (pRBCs) and hemozoin formation were amongst the most common pathological conditions observed in tissues of major affected organs (kidney, liver, lung, brain and spleen) in malarial mice. Neutralization of activin A production via recombinant mouse activin RIIA Fc chimera (rmActivin RIIA Fc chimera) had significantly reduced the parasitemia levels in malarial mice. The release of TNF-α cytokine was significantly reduced as well as the sequestration of parasitized pRBCs and hemozoin formation in major affected organs in malarial mice were also alleviated following inhibition of activin A production. Overall, this preliminary study suggests that activin A could play an immune modulation role in malaria pathogenesis through modulation of TNF-α release that benefits host from severe pathological destructions provoked by intensified inflammatory responses. Further studies are warranted to elucidate the precise mechanism of immune modulation mediated by activin A and its associated immune-modulation mediators in regulating the inflammatory responses elicited during the course of malaria infection.
- Published
- 2021
- Full Text
- View/download PDF
5. Knockdown of dual oxidase 1 suppresses activin A-induced fibrosis in cardiomyocytes via the reactive oxygen species-dependent pyroptotic pathway.
- Author
-
Li S, Li Z, Yin R, Nie J, Fu Y, and Ying R
- Subjects
- Acetylcysteine pharmacology, Activins antagonists & inhibitors, Caspase 1 genetics, Caspase 1 metabolism, Coenzyme A Ligases genetics, Coenzyme A Ligases metabolism, Collagen Type I genetics, Collagen Type I metabolism, Collagen Type III genetics, Collagen Type III metabolism, Dual Oxidases antagonists & inhibitors, Dual Oxidases metabolism, Free Radical Scavengers pharmacology, Gene Expression Regulation, Humans, Interleukin-18 genetics, Interleukin-18 metabolism, Interleukin-1beta genetics, Interleukin-1beta metabolism, Myocytes, Cardiac cytology, Myocytes, Cardiac metabolism, NLR Family, Pyrin Domain-Containing 3 Protein genetics, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Oxidative Stress drug effects, Primary Cell Culture, Pyroptosis genetics, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Reactive Oxygen Species agonists, Reactive Oxygen Species antagonists & inhibitors, Signal Transduction, Smad2 Protein genetics, Smad2 Protein metabolism, Smad3 Protein genetics, Smad3 Protein metabolism, Activins pharmacology, Dual Oxidases genetics, Myocytes, Cardiac drug effects, Pyroptosis drug effects, Reactive Oxygen Species metabolism
- Abstract
Fibrotic diseases account for more than 8 million deaths worldwide annually. Reactive oxygen species (ROS) has been shown to activate pyroptosis and promote the production of interleukin (IL)-1β and IL-18, leading to fibrosis development. However, the role of dual oxidase 1 (DUOX1)-induced ROS production and pyroptosis in cardiac fibrosis remains largely unknown. Activin A was used to induce ROS and pyroptosis in cardiomyocytes. ROS level, pyroptosis, and cytokine production were detected using Active Oxygen Detection Kit, flow cytometry, and enzyme-linked immunosorbent assay, respectively. Western blotting analysis was used to measure expression changes of proteins. DUOX1 was silenced or overexpressed to investigate its role in fibrosis. We found that activin A induced ROS production and pyroptosis in cardiomyocytes, which was blocked by the ROS scavenger, N-acetyl-L-cysteine (NAC). Knockdown of DUOX1 reversed activin A-induced ROS production, pyroptosis, cytokine release, and the upregulation of proinflammatory proteins. Overexpression of DUOX1 resulted in opposite effects of knockdown DUOX1. Administration of an ROS scavenger blocked the effect of DUOX1 overexpression. Supplementation of IL-1β and IL-18 caused significant fibrosis in human cardiac fibroblasts (hCFs). The knockdown of DUOX1 protected cardiomyocytes against activin A-induced fibrosis via the inhibition of ROS, cytokine release, and pyroptosis., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
6. A Two-Step Protocol to Erase Human Skin Fibroblasts and Convert Them into Trophoblast-like Cells.
- Author
-
Arcuri S, Gandolfi F, Somigliana E, and Brevini TAL
- Subjects
- Activins antagonists & inhibitors, Animals, Azacitidine pharmacology, Bone Morphogenetic Protein 4 metabolism, Bone Morphogenetic Protein 4 pharmacology, Cell Differentiation drug effects, Cell Differentiation genetics, Embryo Implantation, Embryo, Mammalian metabolism, Embryonic Stem Cells cytology, Female, Fibroblasts drug effects, Fibroblasts physiology, Humans, MAP Kinase Signaling System drug effects, Mice, Nodal Protein antagonists & inhibitors, Placenta cytology, Pregnancy, Signal Transduction, Skin cytology, Skin growth & development, Cell Culture Techniques methods, Fibroblasts cytology, Trophoblasts cytology
- Abstract
The first differentiation event in mammalian embryos is the formation of the trophectoderm, which is the progenitor of the outer epithelial component of the placenta and supports the fetus during intrauterine life. Our understanding of these events is limited, particularly in human, because of ethical and legal restrictions and availability of adequate in vitro models would be very advantageous. Here we describe a method that converts human fibroblasts into trophoblast-like cells, combining the use of 5-azacytidine-CR (5-aza-CR) to erase the original cell phenotype and a cocktail containing bone morphogenetic protein 4 (BMP4) with inhibitors of the Activin/Nodal/ERK signaling pathways, to drive erased fibroblasts into the trophoblastic differentiation. This innovative method uses very easily accessible cells to derive trophoblast-like cells and it can be useful to study embryo implantation disorders related to aging.
- Published
- 2021
- Full Text
- View/download PDF
7. Pharmacokinetics and Pharmacodynamics of Garetosmab (Anti-Activin A): Results From a First-in-Human Phase 1 Study.
- Author
-
Vanhoutte F, Liang S, Ruddy M, Zhao A, Drewery T, Wang Y, DelGizzi R, Forleo-Neto E, Rajadhyaksha M, Herman G, and Davis JD
- Subjects
- Activins blood, Administration, Intravenous, Antibodies, Neutralizing, Area Under Curve, Double-Blind Method, Female, Healthy Volunteers, Humans, Immunologic Factors administration & dosage, Immunologic Factors blood, Injections, Subcutaneous, Activins antagonists & inhibitors, Immunologic Factors adverse effects, Immunologic Factors pharmacokinetics
- Abstract
We describe outcomes from the first-in-human study of garetosmab (a fully human monoclonal antibody that inhibits activin A) under development for the treatment of fibrodysplasia ossificans progressiva (FOP). In a double-blind, placebo-controlled phase 1 study, 40 healthy women of nonchildbearing potential were randomized to receive a single dose of intravenous garetosmab 0.3, 1, 3, or 10 mg/kg; subcutaneous garetosmab 300 mg; or placebo. Serum concentrations of functional garetosmab (with ≥1 arm free to bind to target), total activin A, and antidrug antibodies were measured predose and up to 113 days post-first dose. Garetosmab demonstrated an acceptable safety profile with no dose-limiting toxicities. Garetosmab displayed nonlinear pharmacokinetics with target-mediated elimination. With increasing doses of intravenous garetosmab, mean peak concentration increased in a dose-proportional manner; mean steady-state estimates ranged from 41.4 to 47.8 mL/kg. A greater than dose-proportional increase in mean area under the concentration-time curve from time zero extrapolated to infinity (range, 72.2-7520 mg*day/L) was observed, consistent with decreasing mean clearance (range, 4.35-1.34 mL/day/kg). Following administration of intravenous garetosmab, mean concentrations of total activin A increased in a dose-dependent manner. At 10 mg/kg, total activin A levels reached a state of little or no change between weeks 4 and 12, suggesting saturation of the target-mediated pathway. No safety signals were seen in this study to preclude investigation in patients. Following intravenous administration, garetosmab concentrations decreased quickly, then decreased over time (reflecting linear elimination), and finally decreased in a nonlinear phase, reflecting target-mediated elimination. Results here support further investigation. Garetosmab 10 mg/kg every 4 weeks intravenously is being evaluated in patients with FOP (NCT03188666)., (© 2020 Regeneron Pharmaceuticals, Inc. The Journal of Clinical Pharmacology published by Wiley Periodicals LLC on behalf of American College of Clinical Pharmacology.)
- Published
- 2020
- Full Text
- View/download PDF
8. Oncogene-Induced Senescence Limits the Progression of Pancreatic Neoplasia through Production of Activin A.
- Author
-
Zhao Y, Wu Z, Chanal M, Guillaumond F, Goehrig D, Bachy S, Principe M, Ziverec A, Flaman JM, Collin G, Tomasini R, Pasternack A, Ritvos O, Vasseur S, Bernard D, Hennino A, and Bertolino P
- Subjects
- Activin Receptors, Type I genetics, Activin Receptors, Type II metabolism, Activins antagonists & inhibitors, Animals, Carcinoma, Pancreatic Ductal metabolism, Disease Progression, Genes, ras, Humans, Mice, Pancreatic Neoplasms metabolism, Phosphorylation, Precancerous Conditions metabolism, Proto-Oncogene Proteins p21(ras) metabolism, Transcriptional Activation, Activin Receptors, Type I metabolism, Activins biosynthesis, Carcinoma, Pancreatic Ductal etiology, Cellular Senescence physiology, Pancreatic Neoplasms etiology, Precancerous Conditions etiology
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a deadly and aggressive cancer. Understanding mechanisms that drive preneoplastic pancreatic lesions is necessary to improve early diagnostic and therapeutic strategies. Mutations and inactivation of activin-like kinase (ALK4) have been demonstrated to favor PDAC onset. Surprisingly, little is known regarding the ligands that drive ALK4 signaling in pancreatic cancer or how this signaling pathway limits the initiation of neoplastic lesions. In this study, data mining and histologic analyses performed on human and mouse tumor tissues revealed that activin A is the major ALK4 ligand that drives PDAC initiation. Activin A, which is absent in normal acinar cells, was strongly induced during acinar-to-ductal metaplasia (ADM), which was promoted by pancreatitis or the activation of Kras
G12D in mice. Activin A expression during ADM was associated with the cellular senescence program that is induced in precursor lesions. Blocking activin A signaling through the use of a soluble form of activin receptor IIB (sActRIIB-Fc) and ALK4 knockout in mice expressing KrasG12D resulted in reduced senescence associated with decreased expression of p21, reduced phosphorylation of H2A histone family member X (H2AX), and increased proliferation. Thus, this study indicates that activin A acts as a protective senescence-associated secretory phenotype factor produced by Kras-induced senescent cells during ADM, which limits the expansion and proliferation of pancreatic neoplastic lesions. SIGNIFICANCE: This study identifies activin A to be a beneficial, senescence-secreted factor induced in pancreatic preneoplastic lesions, which limits their proliferation and ultimately slows progression into pancreatic cancers., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
9. Activin A triggers angiogenesis via regulation of VEGFA and its overexpression is associated with poor prognosis of oral squamous cell carcinoma.
- Author
-
Ervolino De Oliveira C, Dourado MR, Sawazaki-Calone Í, Costa De Medeiros M, Rossa Júnior C, De Karla Cervigne N, Esquiche León J, Lambert D, Salo T, Graner E, and Coletta RD
- Subjects
- Activins analysis, Activins antagonists & inhibitors, Activins genetics, Adult, Aged, Aged, 80 and over, Autocrine Communication drug effects, Autocrine Communication genetics, Cell Movement, Cell Proliferation, Female, Follistatin pharmacology, Follistatin therapeutic use, Gene Knockdown Techniques, Human Umbilical Vein Endothelial Cells, Humans, Male, Middle Aged, Mouth Mucosa pathology, Mouth Neoplasms blood supply, Mouth Neoplasms drug therapy, Mouth Neoplasms mortality, Paracrine Communication drug effects, Paracrine Communication genetics, Phosphorylation drug effects, Phosphorylation genetics, Prognosis, Protein Isoforms metabolism, Smad2 Protein metabolism, Smad3 Protein metabolism, Squamous Cell Carcinoma of Head and Neck blood supply, Squamous Cell Carcinoma of Head and Neck drug therapy, Squamous Cell Carcinoma of Head and Neck mortality, Activins metabolism, Mouth Neoplasms pathology, Neovascularization, Pathologic pathology, Squamous Cell Carcinoma of Head and Neck pathology, Vascular Endothelial Growth Factor A metabolism
- Abstract
Poor prognosis associated with the dysregulated expression of activin A in a number of malignancies has been related to with numerous aspects of tumorigenesis, including angiogenesis. The present study investigated the prognostic significance of activin A immunoexpression in blood vessels and cancer cells in a number of oral squamous cell carcinoma (OSCC) cases and applied in vitro strategies to determine the impact of activin A on angiogenesis. In a cohort of 95 patients with OSCC, immunoexpression of activin A in both blood vessels and tumor cells was quantified and the association with clinicopathological parameters and survival was analyzed. Effects of activin A on the tube formation, proliferation and migration of human umbilical vein endothelial cells (HUVECs) were evaluated in gain‑of‑function (treatment with recombinant activin A) or loss‑of‑function [treatment with activin A‑antagonist follistatin or by stable transfection with short hairpin RNA (shRNA) targeting activin A] conditions. Conditioned medium from an OSCC cell line with shRNA‑mediated depletion of activin A was also tested. The profile of pro‑ and anti‑angiogenic factors regulated by activin A was assessed with a human angiogenesis quantitative PCR (qPCR) array. Vascular endothelial growth factor A (VEGFA) and its major isoforms were evaluated by reverse transcription‑qPCR and ELISA. Activin A expression in blood vessels demonstrated an independent prognostic value in the multivariate analysis with a hazard ratio of 2.47 [95% confidence interval (CI), 1.30‑4.71; P=0.006) for disease‑specific survival and 2.09 (95% CI, 1.07‑4.08l: P=0.03) for disease‑free survival. Activin A significantly increased tubular formation of HUVECs concomitantly with an increase in proliferation. This effect was validated by reduced proliferation and tubular formation of HUVECs following inhibition of activin A by follistatin or shRNA, as well as by treatment of HUVECs with conditioned medium from activin A‑depleted OSCC cells. Activin A‑knockdown increased the migration of HUVECs. In addition, activin A stimulated the phosphorylation of SMAD2/3 and the expression and production of total VEGFA, significantly enhancing the expression of its pro‑angiogenic isoform 121. The present findings suggest that activin A is a predictor of the prognosis of patients with OSCC, and provide evidence that activin A, in an autocrine and paracrine manner, may contribute to OSCC angiogenesis through differential expression of the isoform 121 of VEGFA.
- Published
- 2020
- Full Text
- View/download PDF
10. Activin A Modulates Inflammation in Acute Pancreatitis and Strongly Predicts Severe Disease Independent of Body Mass Index.
- Author
-
Thomas AL, Castellanos K, Mancinelli G, Xia Y, Bauer J, Yazici C, Fantuzzi G, Hwang RF, Krett NL, Papachristou GI, Whitcomb DC, and Jung B
- Subjects
- Activins antagonists & inhibitors, Activins blood, Activins immunology, Animals, Anti-Inflammatory Agents therapeutic use, Biomarkers blood, Biomarkers metabolism, Body Mass Index, Cell Line, Ceruletide administration & dosage, Ceruletide toxicity, Cohort Studies, Disease Models, Animal, Drug Evaluation, Preclinical, Female, Humans, Macrophage Activation immunology, Macrophages, Mice, Pancreas drug effects, Pancreas immunology, Pancreatitis blood, Pancreatitis drug therapy, Pancreatitis immunology, Patient Admission, Predictive Value of Tests, Activins metabolism, Anti-Inflammatory Agents pharmacology, Pancreas pathology, Pancreatitis diagnosis, Severity of Illness Index
- Abstract
Introduction: Acute pancreatitis (AP) is a healthcare challenge with considerable mortality. Treatment is limited to supportive care, highlighting the need to investigate disease drivers and prognostic markers. Activin A is an established mediator of inflammatory responses, and its serum levels correlate with AP severity. We hypothesized that activin A is independent of body mass index (BMI) and is a targetable promoter of the AP inflammatory response., Methods: We assessed whether BMI and serum activin A levels are independent markers to determine disease severity in a cohort of patients with AP. To evaluate activin A inhibition as a therapeutic, we used a cerulein-induced murine model of AP and treated mice with activin A-specific neutralizing antibody or immunoglobulin G control, both before and during the development of AP. We measured the production and release of activin A by pancreas and macrophage cell lines and observed the activation of macrophages after activin A treatment., Results: BMI and activin A independently predicted severe AP in patients. Inhibiting activin A in AP mice reduced disease severity and local immune cell infiltration. Inflammatory stimulation led to activin A production and release by pancreas cells but not by macrophages. Macrophages were activated by activin A, suggesting activin A might promote inflammation in the pancreas in response to injury., Discussion: Activin A provides a promising therapeutic target to interrupt the cycle of inflammation and tissue damage in AP progression. Moreover, assessing activin A and BMI in patients on hospital admission could provide important predictive measures for screening patients likely to develop severe disease.
- Published
- 2020
- Full Text
- View/download PDF
11. Discovery of a follistatin-derived myostatin inhibitory peptide.
- Author
-
Saitoh M, Takayama K, Hitachi K, Taguchi A, Taniguchi A, Tsuchida K, and Hayashi Y
- Subjects
- Activins antagonists & inhibitors, Activins metabolism, Amino Acid Sequence, Animals, Binding Sites, Male, Mice, Mice, Inbred C57BL, Muscle, Skeletal drug effects, Muscle, Skeletal growth & development, Myostatin metabolism, Peptides metabolism, Peptides pharmacology, Protein Binding, Structure-Activity Relationship, Transforming Growth Factor beta antagonists & inhibitors, Transforming Growth Factor beta metabolism, Follistatin chemistry, Myostatin antagonists & inhibitors, Peptides chemistry
- Abstract
Follistatin is well known as an inhibitor of transforming growth factor (TGF)-β superfamily ligands including myostatin and activin A. Myostatin, a negative regulator of muscle growth, is a promising target with which to treat muscle atrophic diseases. Here, we focused on the N-terminal domain (ND) of follistatin (Fst) that interacts with the type I receptor binding site of myostatin. Through bioassay of synthetic ND-derived fragment peptides, we identified DF-3, a new myostatin inhibitory 14-mer peptide which effectively inhibits myostatin, but fails to inhibit activin A or TGF-β1, in an in vitro luciferase reporter assay. Injected intramuscularly, DF-3 significantly increases skeletal muscle mass in mice and consequently, it can serve as a platform for development of muscle enhancement based on myostatin inhibition., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
12. Therapeutic blockade of activin-A improves NK cell function and antitumor immunity.
- Author
-
Rautela J, Dagley LF, de Oliveira CC, Schuster IS, Hediyeh-Zadeh S, Delconte RB, Cursons J, Hennessy R, Hutchinson DS, Harrison C, Kita B, Vivier E, Webb AI, Degli-Esposti MA, Davis MJ, Huntington ND, and Souza-Fonseca-Guimaraes F
- Subjects
- Activins metabolism, Animals, Killer Cells, Natural, Mice, Mice, Knockout, Neoplasm Proteins metabolism, Neoplasms, Experimental metabolism, Neoplasms, Experimental pathology, Activins antagonists & inhibitors, Follistatin pharmacology, Neoplasm Proteins antagonists & inhibitors, Neoplasms, Experimental drug therapy, Signal Transduction drug effects
- Abstract
Natural killer (NK) cells are innate lymphocytes that play a major role in immunosurveillance against tumor initiation and metastatic spread. The signals and checkpoints that regulate NK cell fitness and function in the tumor microenvironment are not well defined. Transforming growth factor-β (TGF-β) is a suppressor of NK cells that inhibits interleukin-15 (IL-15)-dependent signaling events and increases the abundance of receptors that promote tissue residency. Here, we showed that NK cells express the type I activin receptor ALK4, which, upon binding to its ligand activin-A, phosphorylated SMAD2/3 to suppress IL-15-mediated NK cell metabolism. Activin-A impaired human and mouse NK cell proliferation and reduced the production of granzyme B to impair tumor killing. Similar to TGF-β, activin-A also induced SMAD2/3 phosphorylation and stimulated NK cells to increase their cell surface expression of several markers of ILC1 cells. Activin-A also induced these changes in TGF-β receptor-deficient NK cells, suggesting that activin-A and TGF-β stimulate independent pathways that drive SMAD2/3-mediated NK cell suppression. Last, inhibition of activin-A by follistatin substantially slowed orthotopic melanoma growth in mice. These data highlight the relevance of examining TGF-β-independent SMAD2/3 signaling mechanisms as a therapeutic axis to relieve NK cell suppression and promote antitumor immunity., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2019
- Full Text
- View/download PDF
13. Compression of morbidity in a progeroid mouse model through the attenuation of myostatin/activin signalling.
- Author
-
Alyodawi K, Vermeij WP, Omairi S, Kretz O, Hopkinson M, Solagna F, Joch B, Brandt RMC, Barnhoorn S, van Vliet N, Ridwan Y, Essers J, Mitchell R, Morash T, Pasternack A, Ritvos O, Matsakas A, Collins-Hooper H, Huber TB, Hoeijmakers JHJ, and Patel K
- Subjects
- Activin Receptors, Type II administration & dosage, Activin Receptors, Type II genetics, Activins metabolism, Animals, DNA-Binding Proteins genetics, Disease Models, Animal, Endonucleases genetics, Female, Humans, Injections, Intraperitoneal, Male, Mice, Mice, Transgenic, Muscle, Skeletal drug effects, Myostatin metabolism, Recombinant Fusion Proteins administration & dosage, Recombinant Fusion Proteins genetics, Severity of Illness Index, Wasting Syndrome diagnosis, Wasting Syndrome genetics, Wasting Syndrome pathology, Activins antagonists & inhibitors, Aging pathology, Muscle, Skeletal pathology, Signal Transduction drug effects, Wasting Syndrome prevention & control
- Abstract
Background: One of the principles underpinning our understanding of ageing is that DNA damage induces a stress response that shifts cellular resources from growth towards maintenance. A contrasting and seemingly irreconcilable view is that prompting growth of, for example, skeletal muscle confers systemic benefit., Methods: To investigate the robustness of these axioms, we induced muscle growth in a murine progeroid model through the use of activin receptor IIB ligand trap that dampens myostatin/activin signalling. Progeric mice were then investigated for neurological and muscle function as well as cellular profiling of the muscle, kidney, liver, and bone., Results: We show that muscle of Ercc1
Δ/- progeroid mice undergoes severe wasting (decreases in hind limb muscle mass of 40-60% compared with normal mass), which is largely protected by attenuating myostatin/activin signalling using soluble activin receptor type IIB (sActRIIB) (increase of 30-62% compared with untreated progeric). sActRIIB-treated progeroid mice maintained muscle activity (distance travel per hour: 5.6 m in untreated mice vs. 13.7 m in treated) and increased specific force (19.3 mN/mg in untreated vs. 24.0 mN/mg in treated). sActRIIb treatment of progeroid mice also improved satellite cell function especially their ability to proliferate on their native substrate (2.5 cells per fibre in untreated progeroids vs. 5.4 in sActRIIB-treated progeroids after 72 h in culture). Besides direct protective effects on muscle, we show systemic improvements to other organs including the structure and function of the kidneys; there was a major decrease in the protein content in urine (albumin/creatinine of 4.9 sActRIIB treated vs. 15.7 in untreated), which is likely to be a result in the normalization of podocyte foot processes, which constitute the filtration apparatus (glomerular basement membrane thickness reduced from 224 to 177 nm following sActRIIB treatment). Treatment of the progeric mice with the activin ligand trap protected against the development of liver abnormalities including polyploidy (18.3% untreated vs. 8.1% treated) and osteoporosis (trabecular bone volume; 0.30 mm3 in treated progeroid mice vs. 0.14 mm3 in untreated mice, cortical bone volume; 0.30 mm3 in treated progeroid mice vs. 0.22 mm3 in untreated mice). The onset of neurological abnormalities was delayed (by ~5 weeks) and their severity reduced, overall sustaining health without affecting lifespan., Conclusions: This study questions the notion that tissue growth and maintaining tissue function during ageing are incompatible mechanisms. It highlights the need for future investigations to assess the potential of therapies based on myostatin/activin blockade to compress morbidity and promote healthy ageing., (© 2019 The Authors. Journal of Cachexia, Sarcopenia and Muscle published by John Wiley & Sons Ltd on behalf of the Society on Sarcopenia, Cachexia and Wasting Disorders.)- Published
- 2019
- Full Text
- View/download PDF
14. Myostatin and activin blockade by engineered follistatin results in hypertrophy and improves dystrophic pathology in mdx mouse more than myostatin blockade alone.
- Author
-
Iskenderian A, Liu N, Deng Q, Huang Y, Shen C, Palmieri K, Crooker R, Lundberg D, Kastrapeli N, Pescatore B, Romashko A, Dumas J, Comeau R, Norton A, Pan J, Rong H, Derakhchan K, and Ehmann DE
- Subjects
- Animals, Follistatin administration & dosage, Hand Strength, Male, Mice, Mice, Inbred C57BL, Mice, Inbred mdx, Muscle Contraction, Recombinant Proteins administration & dosage, Recombinant Proteins therapeutic use, Activins antagonists & inhibitors, Follistatin therapeutic use, Muscular Dystrophies drug therapy, Myostatin antagonists & inhibitors
- Abstract
Background: Myostatin antagonists are being developed as therapies for Duchenne muscular dystrophy due to their strong hypertrophic effects on skeletal muscle. Engineered follistatin has the potential to combine the hypertrophy of myostatin antagonism with the anti-inflammatory and anti-fibrotic effects of activin A antagonism., Methods: Engineered follistatin was administered to C57BL/6 mice for 4 weeks, and muscle mass and myofiber size was measured. In the mdx model, engineered follistatin was dosed for 12 weeks in two studies comparing to an Fc fusion of the activin IIB receptor or an anti-myostatin antibody. Functional measurements of grip strength and tetanic force were combined with tissue analysis for markers of necrosis, inflammation, and fibrosis to evaluate improvement in dystrophic pathology., Results: In wild-type and mdx mice, dose-dependent increases in muscle mass and quadriceps myofiber size were observed for engineered follistatin. In mdx, increases in grip strength and tetanic force were combined with improvements in muscle markers for necrosis, inflammation, and fibrosis. Improvements in dystrophic pathology were greater for engineered follistatin than the anti-myostatin antibody., Conclusions: Engineered follistatin generated hypertrophy and anti-fibrotic effects in the mdx model.
- Published
- 2018
- Full Text
- View/download PDF
15. The regulatory protein SnoN antagonizes activin/Smad2 protein signaling and thereby promotes adipocyte differentiation and obesity in mice.
- Author
-
Zhu Q, Chang A, Xu A, and Luo K
- Subjects
- Activins antagonists & inhibitors, Activins metabolism, Adipose Tissue, White growth & development, Animals, Mesenchymal Stem Cells cytology, Mice, Smad2 Protein antagonists & inhibitors, Adipocytes cytology, Cell Differentiation, Obesity, Proto-Oncogene Proteins physiology, Signal Transduction
- Abstract
Ski-related oncogene SnoN (SnoN or SKIL) regulates multiple signaling pathways in a tissue- and developmental stage-dependent manner and has broad functions in embryonic angiogenesis, mammary gland alveologenesis, cancer, and aging. Here, we report that SnoN also plays a critical role in white adipose tissue (WAT) development by regulating mesenchymal stem cell (MSC) self-renewal and differentiation. We found that SnoN promotes MSC differentiation in the adipocyte lineage by antagonizing activin A/Smad2, but not TGFβ/Smad3 signaling. Mice lacking SnoN or expressing a mutant SnoN defective in binding to the Smads were protected from high-fat diet-induced obesity and insulin resistance, and MSCs lacking a functional SnoN exhibited defective differentiation. We further demonstrated that activin, via Smad2, appears to be the major regulator of WAT development in vivo We also noted that activin A is abundantly expressed in WAT and adipocytes through an autocrine mechanism and promotes MSC self-renewal and inhibits adipogenic differentiation by inducing expression of the gene encoding the homeobox transcription factor Nanog. Of note, SnoN repressed activin/Smad2 signaling and activin A expression, enabling expression of adipocyte-specific transcription factors and promoting adipogenic differentiation. In conclusion, our study has revealed that SnoN plays an important in vivo role in adipocyte differentiation and WAT development in vivo by decreasing activity in the activin/Smad2 signaling pathway.
- Published
- 2018
- Full Text
- View/download PDF
16. Follistatin Expression in Human Invasive Breast Tumors: Pathologic and Clinical Associations.
- Author
-
Couto HL, Dela Cruz C, Buzelin MA, Toppa NH, Wainstein AJ, and Reis FM
- Subjects
- Activins antagonists & inhibitors, Breast Neoplasms pathology, Carcinogenesis, Cell Cycle, Cell Proliferation, Disease Progression, Estrogen Receptor alpha metabolism, Female, Follow-Up Studies, Humans, Immunohistochemistry, Menopause, Neoplasm Metastasis, Neoplasm Staging, Paraffin Embedding, Receptor, ErbB-2 metabolism, Breast Neoplasms metabolism, Follistatin metabolism, Mammary Glands, Human physiology
- Abstract
Follistatin is a potent native activin antagonist that is expressed in the normal mammary gland and in different breast proliferative diseases. Despite experimental evidence that follistatin can modulate the breast cancer cell cycle, the clinical significance of follistatin expression in these tumors is unknown. The aim of this study was to correlate the intensity of follistatin expression in invasive breast cancer with some of its clinical and pathologic features, such as the disease stage and the hormonal receptor status. Paraffin blocks of tumor samples that had been fixed in buffered formalin were obtained from 154 women subjected to surgery for breast cancer between 2008 and 2012. Sections from all paraffin blocks were cut and processed together by immunohistochemistry using a commercial monoclonal antibody to human follistatin. The intensity of follistatin staining was unrelated to the menopausal status, the disease stage, the grade, progesterone receptor expression, and local or systemic recurrence. However, follistatin immunoreactivity was significantly stronger in estrogen receptor (ER)-negative tumors than in ER-positive tumors. These findings suggest that follistatin expression in invasive breast cancer is unrelated to the disease severity and the risk of recurrence, but is more intense in ER-negative tumors.
- Published
- 2018
- Full Text
- View/download PDF
17. Induction of experimental autoimmune orchitis in mice: responses to elevated circulating levels of the activin-binding protein, follistatin.
- Author
-
Nicolas N, Muir JA, Hayward S, Chen JL, Stanton PG, Gregorevic P, de Kretser DM, Loveland KL, Bhushan S, Meinhardt A, Fijak M, and Hedger MP
- Subjects
- Activins antagonists & inhibitors, Activins metabolism, Animals, Autoimmune Diseases immunology, Autoimmune Diseases metabolism, Autoimmune Diseases pathology, Biomarkers blood, Biomarkers metabolism, Blood-Testis Barrier immunology, Blood-Testis Barrier metabolism, Blood-Testis Barrier pathology, Blood-Testis Barrier physiopathology, Disease Progression, Fibrosis, Follistatin administration & dosage, Follistatin genetics, Follistatin metabolism, Gene Expression Regulation, Gene Transfer Techniques, Inflammation Mediators metabolism, Male, Mice, Inbred C57BL, Orchitis immunology, Orchitis metabolism, Orchitis pathology, Recombinant Proteins administration & dosage, Recombinant Proteins blood, Recombinant Proteins metabolism, Testis immunology, Testis pathology, Apoptosis, Autoimmune Diseases physiopathology, Disease Models, Animal, Follistatin blood, Orchitis physiopathology, Testis metabolism, Up-Regulation
- Abstract
Experimental autoimmune orchitis (EAO) is a rodent model of chronic testicular inflammation that mimics the pathology observed in some types of human infertility. In a previous study, testicular expression of the inflammatory/immunoregulatory cytokine, activin A, was elevated in adult mice during the onset of EAO, indicating a potential role in the regulation of the disease. Consequently, we examined the development of EAO in mice with elevated levels of follistatin, an endogenous activin antagonist, as a potential therapeutic approach to testicular inflammation. Prior to EAO induction, mice received a single intramuscular injection of a non-replicative recombinant adeno-associated viral vector carrying a gene cassette of the circulating form of follistatin, FST315 (FST group). Serum follistatin levels were increased 5-fold in the FST group compared with the control empty vector (EV) group at 30 and 50 days of EAO, but intra-testicular levels of follistatin or activin A were not significantly altered. Induction of EAO was reduced, but not prevented, with mild-to-severe damage in 75% of the EV group and 40% of the FST group, at 50 days following immunisation with testicular homogenate. However, the EAO damage score (based on disruption of the blood-testis barrier, apoptosis, testicular damage and fibrosis) and extent of intratesticular inflammation (expression of inflammatory mediators) were directly proportional to the levels of activin A measured in the testis at 50 days. These data implicate activin A in the progression of EAO, thereby providing a potential therapeutic target; however, elevating circulating follistatin levels were not sufficient to prevent EAO development., (© 2017 Society for Reproduction and Fertility.)
- Published
- 2017
- Full Text
- View/download PDF
18. Specific targeting of TGF-β family ligands demonstrates distinct roles in the regulation of muscle mass in health and disease.
- Author
-
Chen JL, Walton KL, Hagg A, Colgan TD, Johnson K, Qian H, Gregorevic P, and Harrison CA
- Subjects
- Activins antagonists & inhibitors, Activins genetics, Activins metabolism, Animals, Gene Targeting, Male, Mice, Muscle, Skeletal pathology, Organ Size genetics, Smad Proteins genetics, Smad Proteins metabolism, Dependovirus, Genetic Vectors, Muscle Proteins antagonists & inhibitors, Muscle Proteins genetics, Muscle Proteins metabolism, Muscle, Skeletal growth & development, Muscular Diseases genetics, Muscular Diseases metabolism, Muscular Diseases pathology, Signal Transduction, Transforming Growth Factor beta antagonists & inhibitors, Transforming Growth Factor beta genetics, Transforming Growth Factor beta metabolism
- Abstract
The transforming growth factor-β (TGF-β) network of ligands and intracellular signaling proteins is a subject of intense interest within the field of skeletal muscle biology. To define the relative contribution of endogenous TGF-β proteins to the negative regulation of muscle mass via their activation of the Smad2/3 signaling axis, we used local injection of adeno-associated viral vectors (AAVs) encoding ligand-specific antagonists into the tibialis anterior (TA) muscles of C57BL/6 mice. Eight weeks after AAV injection, inhibition of activin A and activin B signaling produced moderate (∼20%), but significant, increases in TA mass, indicating that endogenous activins repress muscle growth. Inhibiting myostatin induced a more profound increase in muscle mass (∼45%), demonstrating a more prominent role for this ligand as a negative regulator of adult muscle mass. Remarkably, codelivery of activin and myostatin inhibitors induced a synergistic response, resulting in muscle mass increasing by as much as 150%. Transcription and protein analysis indicated that this substantial hypertrophy was associated with both the complete inhibition of the Smad2/3 pathway and activation of the parallel bone morphogenetic protein (BMP)/Smad1/5 axis (recently identified as a positive regulator of muscle mass). Analyses indicated that hypertrophy was primarily driven by an increase in protein synthesis, but a reduction in ubiquitin-dependent protein degradation pathways was also observed. In models of muscular dystrophy and cancer cachexia, combined inhibition of activins and myostatin increased mass or prevented muscle wasting, respectively, highlighting the potential therapeutic advantages of specifically targeting multiple Smad2/3-activating ligands in skeletal muscle., Competing Interests: The authors declare no conflict of interest.
- Published
- 2017
- Full Text
- View/download PDF
19. Impairment of the activin A autocrine loop by lopinavir reduces self-renewal of distinct human adipose progenitors.
- Author
-
Ravaud C, Paré M, Azoulay S, Dani C, and Ladoux A
- Subjects
- Cells, Cultured, Gene Expression Regulation drug effects, Humans, Protein Interaction Maps drug effects, Stem Cells drug effects, Activins antagonists & inhibitors, Adipose Tissue cytology, Cell Proliferation drug effects, HIV Protease Inhibitors adverse effects, Lopinavir adverse effects, Stem Cells physiology
- Abstract
Maintenance of the adipose tissue requires a proper balance between self-renewal and differentiation of adipose progenitors (AP). Any deregulation leads either to fat overexpansion and obesity or fat loss and consequent lipodystrophies. Depending on the fat pad location, APs and adipocytes are heterogeneous. However, information on the pharmacological sensitivity of distinct APs to drugs known to alter the function of adipose tissue, especially HIV protease inhibitors (PIs) is scant. Here we show that PIs decreased proliferation and clonal expansion of APs, modifying their self-renewal potential. Lopinavir was the most potent PI tested. Decrease in self-renewal was accompanied by a reduced expression of the immediate early response gene IER3, a gene associated with tissue expansion. It was more pronounced in chin-derived APs than in knee-derived APs. Furthermore, lopinavir lowered the activin A-induced ERK1/2 phosphorylation. Expressions of the transcription factor EGR1 and its targets, including INHBA were subsequently altered. Therefore, activin A secretion was reduced leading to a dramatic impairment of APs self-renewal sustained by the activin A autocrine loop. All together, these observations highlight the activin A autocrine loop as a crucial effector to maintain APs self-renewal. Targeting this pathway by HIV-PIs may participate in the induction of unwanted side effects.
- Published
- 2017
- Full Text
- View/download PDF
20. Activin A more prominently regulates muscle mass in primates than does GDF8.
- Author
-
Latres E, Mastaitis J, Fury W, Miloscio L, Trejos J, Pangilinan J, Okamoto H, Cavino K, Na E, Papatheodorou A, Willer T, Bai Y, Hae Kim J, Rafique A, Jaspers S, Stitt T, Murphy AJ, Yancopoulos GD, and Gromada J
- Subjects
- Activin Receptors, Type II metabolism, Activins antagonists & inhibitors, Animals, Antibodies, Monoclonal pharmacology, Body Mass Index, Dexamethasone pharmacology, Humans, Isometric Contraction physiology, Macaca fascicularis, Male, Mice, Mice, Inbred C57BL, Mice, SCID, Muscle, Skeletal physiology, Myostatin antagonists & inhibitors, Rats, Activins metabolism, Hypertrophy pathology, Muscle Hypotonia pathology, Muscle, Skeletal growth & development, Myostatin metabolism
- Abstract
Growth and differentiation factor 8 (GDF8) is a TGF-β superfamily member, and negative regulator of skeletal muscle mass. GDF8 inhibition results in prominent muscle growth in mice, but less impressive hypertrophy in primates, including man. Broad TGF-β inhibition suggests another family member negatively regulates muscle mass, and its blockade enhances muscle growth seen with GDF8-specific inhibition. Here we show that activin A is the long-sought second negative muscle regulator. Activin A specific inhibition, on top of GDF8 inhibition, leads to pronounced muscle hypertrophy and force production in mice and monkeys. Inhibition of these two ligands mimics the hypertrophy seen with broad TGF-β blockers, while avoiding the adverse effects due to inhibition of multiple family members. Altogether, we identify activin A as a second negative regulator of muscle mass, and suggest that inhibition of both ligands provides a preferred therapeutic approach, which maximizes the benefit:risk ratio for muscle diseases in man.
- Published
- 2017
- Full Text
- View/download PDF
21. Directing Differentiation of Pluripotent Stem Cells Toward Retinal Pigment Epithelium Lineage.
- Author
-
Choudhary P, Booth H, Gutteridge A, Surmacz B, Louca I, Steer J, Kerby J, and Whiting PJ
- Subjects
- Activins antagonists & inhibitors, Activins metabolism, Bone Morphogenetic Proteins antagonists & inhibitors, Bone Morphogenetic Proteins metabolism, Cells, Cultured, Epithelial Cells drug effects, Epithelial Cells metabolism, Gene Expression Regulation, Developmental, Humans, Induced Pluripotent Stem Cells drug effects, Induced Pluripotent Stem Cells metabolism, Phenotype, Pyrazoles pharmacology, Pyrimidines pharmacology, Retinal Pigment Epithelium drug effects, Retinal Pigment Epithelium metabolism, Signal Transduction, Time Factors, Transcriptome, Cell Differentiation drug effects, Cell Lineage drug effects, Cellular Reprogramming, Cellular Reprogramming Techniques, Epithelial Cells physiology, Induced Pluripotent Stem Cells physiology, Retinal Pigment Epithelium physiology
- Abstract
Development of efficient and reproducible conditions for directed differentiation of pluripotent stem cells into specific cell types is important not only to understand early human development but also to enable more practical applications, such as in vitro disease modeling, drug discovery, and cell therapies. The differentiation of stem cells to retinal pigment epithelium (RPE) in particular holds promise as a source of cells for therapeutic replacement in age-related macular degeneration. Here we show development of an efficient method for deriving homogeneous RPE populations in a period of 45 days using an adherent, monolayer system and defined xeno-free media and matrices. The method utilizes sequential inhibition and activation of the Activin and bone morphogenetic protein signaling pathways and can be applied to both human embryonic stem cells and induced pluripotent stem cells as the starting population. In addition, we use whole genome transcript analysis to characterize cells at different stages of differentiation that provides further understanding of the developmental dynamics and fate specification of RPE. We show that with the described method, RPE develop through stages consistent with their formation during embryonic development. This characterization- together with the absence of steps involving embryoid bodies, three-dimensional culture, or manual dissections, which are common features of other protocols-makes this process very attractive for use in research as well as for clinical applications. Stem Cells Translational Medicine 2017;6:490-501., (© 2016 The Authors Stem Cells Translational Medicine published by Wiley Periodicals, Inc. on behalf of AlphaMed Press.)
- Published
- 2017
- Full Text
- View/download PDF
22. Activin inhibition limits early innate immune response in rat kidney allografts-a pilot study.
- Author
-
Palin NK, Savikko J, Pasternack A, Rintala JM, Kalra B, Mistry S, Kumar A, Roth MP, Helin H, and Ritvos O
- Subjects
- Activins metabolism, Animals, Cytokines metabolism, Enzyme-Linked Immunosorbent Assay, Fibroblasts metabolism, Humans, Inflammation, Male, Pilot Projects, Rats, Rats, Wistar, Renal Insufficiency surgery, Reperfusion Injury, Signal Transduction, Time Factors, Transforming Growth Factor beta metabolism, Transplantation, Homologous, Activins antagonists & inhibitors, Allografts immunology, Immunity, Innate, Kidney immunology, Kidney Transplantation
- Abstract
Activins are members of the transforming growth factor-beta (TGF-β) superfamily of cytokines. They play critical roles in the onset of acute and chronic inflammatory responses. The aim of this study was to investigate how activin inhibition affects acute kidney injury and inflammation after transplantation. The study was carried out in kidney transplantation and renal ischemia-reperfusion models in the rat. Soluble activin type 2 receptor (sActRIIB-Fc) was used to inhibit activin signaling. Transplantation groups were as follows: (i) cyclosporine A (CsA) (ii) CsA + sActRIIB-Fc, (iii) CsA+ inactive protein control Fc-G1. IRI groups were as follows: (i) no treatment, (ii) sActRIIB-Fc. Serum activin B concentration was significantly elevated after transplantation and IRI, whereas activin A was produced locally in renal allografts. Activin inhibition efficiently limited neutrophil, macrophage, and dendritic cell infiltration to the allografts measured 72 h after transplantation. In addition, sActRIIB-Fc treatment modulated serum cytokine response after transplantation and reduced the early accumulation of fibroblasts in the graft interstitium. In conclusion activin inhibition reduces the innate immune response early after renal transplantation in the rat. It also limits the accumulation of fibroblasts in the graft suggesting that activins may be involved in the fibrogenic signaling already early after kidney transplantation., (© 2016 Steunstichting ESOT.)
- Published
- 2017
- Full Text
- View/download PDF
23. Inhibition of Activin Signaling Slows Progression of Polycystic Kidney Disease.
- Author
-
Leonhard WN, Kunnen SJ, Plugge AJ, Pasternack A, Jianu SB, Veraar K, El Bouazzaoui F, Hoogaars WM, Ten Dijke P, Breuning MH, De Heer E, Ritvos O, and Peters DJ
- Subjects
- Animals, Disease Progression, Epithelial Cells, Female, Kidney cytology, Male, Mice, Polycystic Kidney Diseases etiology, Recombinant Fusion Proteins pharmacology, Smad2 Protein physiology, Smad3 Protein physiology, Time Factors, Activins antagonists & inhibitors, Polycystic Kidney Diseases prevention & control, Signal Transduction
- Abstract
Autosomal dominant polycystic kidney disease (ADPKD), characterized by the formation of numerous kidney cysts, is caused by PKD1 or PKD2 mutations and affects 0.1% of the population. Although recent clinical studies indicate that reduction of cAMP levels slows progression of PKD, this finding has not led to an established safe and effective therapy for patients, indicating the need to find new therapeutic targets. The role of TGF-β in PKD is not clearly understood, but nuclear accumulation of phosphorylated SMAD2/3 in cyst-lining cells suggests the involvement of TGF-β signaling in this disease. In this study, we ablated the TGF-β type 1 receptor (also termed activin receptor-like kinase 5) in renal epithelial cells of PKD mice, which had little to no effect on the expression of SMAD2/3 target genes or the progression of PKD. Therefore, we investigated whether alternative TGF-β superfamily ligands account for SMAD2/3 activation in cystic epithelial cells. Activins are members of the TGF-β superfamily and drive SMAD2/3 phosphorylation via activin receptors, but activins have not been studied in the context of PKD. Mice with PKD had increased expression of activin ligands, even at early stages of disease. In addition, treatment with a soluble activin receptor IIB fusion (sActRIIB-Fc) protein, which acts as a soluble trap to sequester activin ligands, effectively inhibited cyst formation in three distinct mouse models of PKD. These data point to activin signaling as a key pathway in PKD and a promising target for therapy., (Copyright © 2016 by the American Society of Nephrology.)
- Published
- 2016
- Full Text
- View/download PDF
24. Transforming Growth Factor-β Family Ligands Can Function as Antagonists by Competing for Type II Receptor Binding.
- Author
-
Aykul S and Martinez-Hackert E
- Subjects
- Activins antagonists & inhibitors, Activins metabolism, Activins pharmacology, Binding, Competitive, Bone Morphogenetic Protein 2 metabolism, Bone Morphogenetic Protein 7 metabolism, Bone Morphogenetic Proteins metabolism, Cell Line, Tumor, Follistatin pharmacology, Growth Differentiation Factor 2, Growth Differentiation Factors metabolism, Hep G2 Cells, Humans, Ligands, Recombinant Proteins metabolism, Signal Transduction drug effects, Activin Receptors, Type II antagonists & inhibitors, Activin Receptors, Type II metabolism, Bone Morphogenetic Protein Receptors, Type II antagonists & inhibitors, Bone Morphogenetic Protein Receptors, Type II metabolism, Transforming Growth Factor beta metabolism
- Abstract
Transforming growth factor-β (TGF-β) family ligands are pleiotropic cytokines. Their physiological activities are not determined by a simple coupling of stimulus and response, but depend critically on context, i.e. the interplay of receptors, ligands, and regulators that form the TGF-β signal transduction system of a cell or tissue. How these different components combine to regulate signaling activities remains poorly understood. Here, we describe a ligand-mediated mechanism of signaling regulation. Based on the observation that the type II TGF-β family receptors ActRIIA, ActRIIB, and BMPRII interact with a large group of overlapping ligands at overlapping epitopes, we hypothesized high affinity ligands compete with low affinity ligands for receptor binding and signaling. We show activin A and other high affinity ligands directly inhibited signaling by the low affinity ligands BMP-2, BMP-7, and BMP-9. We demonstrate activin A functions as a competitive inhibitor that blocks the ligand binding epitope on type II receptors. We propose binding competition and signaling antagonism are integral functions of the TGF-β signal transduction system. These functions could help explain how activin A modulates physiological signaling during extraordinary cellular responses, such as injury and wound healing, and how activin A could elicit disease phenotypes such as cancer-related muscle wasting and fibrosis., (© 2016 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
25. New expectations in the treatment of anemia in chronic kidney disease.
- Author
-
López-Gómez JM, Abad S, and Vega A
- Subjects
- Activins antagonists & inhibitors, Anemia etiology, Basic Helix-Loop-Helix Transcription Factors physiology, Clinical Trials as Topic, Drugs, Investigational therapeutic use, Erythropoiesis drug effects, Erythropoietin physiology, GATA Transcription Factors antagonists & inhibitors, GATA Transcription Factors physiology, Hematinics classification, Hematinics pharmacology, Hepcidins antagonists & inhibitors, Humans, Multicenter Studies as Topic, Prolyl-Hydroxylase Inhibitors pharmacology, Prolyl-Hydroxylase Inhibitors therapeutic use, Randomized Controlled Trials as Topic, Receptors, Erythropoietin agonists, Renal Dialysis adverse effects, Renal Insufficiency, Chronic blood, Renal Insufficiency, Chronic therapy, Anemia drug therapy, Hematinics therapeutic use, Renal Insufficiency, Chronic complications
- Abstract
The new drugs developed for the treatment of anemia in chronic kidney disease patients, together with their mechanisms of action are reviewed. At present, many of them are already in advanced stages of clinical trials and is expected to be incorporated into the therapeutic arsenal in the coming years. The potential benefits and possible limitations are also described., (Copyright © 2016 Sociedad Española de Nefrología. Published by Elsevier España, S.L.U. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
26. Role of activin A in the pathogenesis of endothelial cell dysfunction in preeclampsia.
- Author
-
Hobson SR, Acharya R, Lim R, Chan ST, Mockler J, and Wallace EM
- Subjects
- Activins antagonists & inhibitors, Adult, Biomarkers metabolism, Case-Control Studies, Female, Human Umbilical Vein Endothelial Cells physiology, Humans, Pre-Eclampsia blood, Pre-Eclampsia physiopathology, Pregnancy, Up-Regulation, Activins metabolism, Endothelin-1 metabolism, Human Umbilical Vein Endothelial Cells metabolism, Intercellular Adhesion Molecule-1 metabolism, Pre-Eclampsia etiology, Vascular Cell Adhesion Molecule-1 metabolism
- Abstract
Circulating markers for endothelial activation such as endothelin-1 (ET-1), ICAM-1 and VCAM-1 are elevated in women with preeclampsia. Using human umbilical vein endothelial cells (HUVECs) as an in vitro model of the maternal vasculature, we show that activin A and preeclamptic serum upregulate ET-1, ICAM-1, and VCAM-1 in HUVECs. Further, we show that follistatin, a specific binding protein for activin, mitigates the upregulation of ET-1, ICAM-1 and VCAM-1 in HUVECs exposed to either activin A or preeclamptic serum. These data are consistent with activin A contributing to the pathophysiology of preeclampsia and suggest that therapies targeting activin signalling are worth exploring., (Copyright © 2016 International Society for the Study of Hypertension in Pregnancy. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
27. Activin A Increases Human Trophoblast Invasion by Inducing SNAIL-Mediated MMP2 Up-Regulation Through ALK4.
- Author
-
Li Y, Klausen C, Zhu H, and Leung PC
- Subjects
- Activin Receptors, Type I antagonists & inhibitors, Activin Receptors, Type I chemistry, Activin Receptors, Type I genetics, Activins antagonists & inhibitors, Adult, Cell Line, Transformed, Cell Movement, Cells, Cultured, Female, Humans, Matrix Metalloproteinase 2 chemistry, Matrix Metalloproteinase 2 genetics, Pregnancy, Pregnancy Trimester, First, RNA Interference, RNA, Messenger metabolism, Signal Transduction, Smad Proteins agonists, Smad Proteins antagonists & inhibitors, Smad Proteins genetics, Smad Proteins metabolism, Snail Family Transcription Factors, Transcription Factors antagonists & inhibitors, Transcription Factors genetics, Transcription Factors metabolism, Trophoblasts cytology, Activin Receptors, Type I metabolism, Activins metabolism, Matrix Metalloproteinase 2 metabolism, Placentation, Transcription Factors agonists, Trophoblasts metabolism, Up-Regulation
- Abstract
Context: Activin A increases matrix metalloproteinase (MMP) 2 expression and cell invasion in human trophoblasts, but whether the expression of MMP2 is essential for the proinvasive effect of activin A has yet to be determined. Moreover, the identity of the activin receptor-like kinase (ALK; TGF-β type I receptors) and downstream transcription factors (eg, SNAIL and SLUG) mediating the effects of activin on MMP2 expression and trophoblast cell invasion remains unknown., Objective: To elucidate the role of MMP2 in activin A-induced human trophoblast cell invasion as well as the involvement of ALK4 and SNAIL., Design: HTR8/SVneo immortalized human extravillous cytotrophoblast (EVT) cells and primary cultures of human first-trimester EVT cells were used as study models. Small interfering RNA (siRNA)-mediated knockdown approaches were used to investigate the molecular determinants of activin A-mediated functions., Main Outcome Measures: Levels of mRNA and protein were examined by reverse transcription-quantitative real-time PCR and Western blot, respectively. Cell invasiveness was measured by Matrigel-coated transwell assays., Results: Treatment of HTR8/SVneo cells with activin A increased the production of SNAIL, SLUG, and MMP2 without altering that of MMP9, TIMP1, TIMP2, TWIST, RUNX2, ZEB1, or ZEB2. Similarly, activin A up-regulated the mRNA and protein levels of SNAIL and MMP2 in primary EVT cells. Knockdown of SNAIL attenuated activin A-induced MMP2 up-regulation in HTR8/SVneo and primary EVT cells. In HTR8/SVneo cells, activin A-induced production of SNAIL and MMP2 was abolished by pretreatment with the TGF-β type I receptor (ALK4/5/7) inhibitor SB431542 or siRNA targeting ALK4, SMAD2/3, or common SMAD4. Likewise, knockdown of ALK4 or SMAD4 abolished the stimulatory effects of activin A on SNAIL and MMP2 expression in primary EVT cells. Importantly, activin A-induced HTR8/SVneo and primary EVT cell invasion were attenuated by siRNA-mediated depletion of ALK4 or MMP2., Conclusion: Activin A induces human trophoblast cell invasion by inducing SNAIL-mediated MMP2 expression through ALK4 in a SMAD2/3-SMAD4-dependent manner.
- Published
- 2015
- Full Text
- View/download PDF
28. Virtual High-Throughput Screening To Identify Novel Activin Antagonists.
- Author
-
Zhu J, Mishra RK, Schiltz GE, Makanji Y, Scheidt KA, Mazar AP, and Woodruff TK
- Subjects
- Activins chemistry, Activins metabolism, Animals, Cell Line, Tumor, Cell Proliferation drug effects, Crystallography, X-Ray, Drug Evaluation, Preclinical, Female, Follicle Stimulating Hormone antagonists & inhibitors, Hep G2 Cells, Humans, Mice, Molecular Docking Simulation, Ovary cytology, Ovary drug effects, Protein Conformation, Signal Transduction drug effects, Activins antagonists & inhibitors, High-Throughput Screening Assays, User-Computer Interface
- Abstract
Activin belongs to the TGFβ superfamily, which is associated with several disease conditions, including cancer-related cachexia, preterm labor with delivery, and osteoporosis. Targeting activin and its related signaling pathways holds promise as a therapeutic approach to these diseases. A small-molecule ligand-binding groove was identified in the interface between the two activin βA subunits and was used for a virtual high-throughput in silico screening of the ZINC database to identify hits. Thirty-nine compounds without significant toxicity were tested in two well-established activin assays: FSHβ transcription and HepG2 cell apoptosis. This screening workflow resulted in two lead compounds: NUCC-474 and NUCC-555. These potential activin antagonists were then shown to inhibit activin A-mediated cell proliferation in ex vivo ovary cultures. In vivo testing showed that our most potent compound (NUCC-555) caused a dose-dependent decrease in FSH levels in ovariectomized mice. The Blitz competition binding assay confirmed target binding of NUCC-555 to the activin A:ActRII that disrupts the activin A:ActRII complex's binding with ALK4-ECD-Fc in a dose-dependent manner. The NUCC-555 also specifically binds to activin A compared with other TGFβ superfamily member myostatin (GDF8). These data demonstrate a new in silico-based strategy for identifying small-molecule activin antagonists. Our approach is the first to identify a first-in-class small-molecule antagonist of activin binding to ALK4, which opens a completely new approach to inhibiting the activity of TGFβ receptor superfamily members. in addition, the lead compound can serve as a starting point for lead optimization toward the goal of a compound that may be effective in activin-mediated diseases.
- Published
- 2015
- Full Text
- View/download PDF
29. The activin A antagonist follistatin inhibits cystic fibrosis-like lung inflammation and pathology.
- Author
-
Hardy CL, King SJ, Mifsud NA, Hedger MP, Phillips DJ, Mackay F, de Kretser DM, Wilson JW, Rolland JM, and O'Hehir RE
- Subjects
- Activins blood, Adult, Animals, Body Weight drug effects, Cystic Fibrosis genetics, Cystic Fibrosis metabolism, Cystic Fibrosis physiopathology, Disease Models, Animal, Female, Follistatin pharmacology, Humans, Inflammation Mediators metabolism, Lung drug effects, Lung metabolism, Lung pathology, Macrophages immunology, Macrophages pathology, Male, Mice, Mice, Transgenic, Middle Aged, Mucus metabolism, Neutrophil Infiltration, Neutrophils immunology, Neutrophils pathology, Pneumonia drug therapy, Pneumonia pathology, Pneumonia physiopathology, Respiratory Function Tests, Respiratory Mucosa immunology, Respiratory Mucosa metabolism, Respiratory Mucosa pathology, Young Adult, Activins antagonists & inhibitors, Cystic Fibrosis complications, Follistatin metabolism, Pneumonia etiology, Pneumonia metabolism
- Abstract
Cystic fibrosis (CF) is the most common life-limiting genetically acquired respiratory disorder. Patients with CF have thick mucus obstructing the airways leading to recurrent infections, bronchiectasis and neutrophilic airway inflammation culminating in deteriorating lung function. Current management targets airway infection and mucus clearance, but despite recent advances in care, life expectancy is still only 40 years. We investigated whether activin A is elevated in CF lung disease and whether inhibiting activin A with its natural antagonist follistatin retards lung disease progression. We measured serum activin A levels, lung function and nutritional status in CF patients. We studied the effect of activin A on CF lung pathogenesis by treating newborn CF transgenic mice (β-ENaC) intranasally with the natural activin A antagonist follistatin. Activin A levels were elevated in the serum of adult CF patients, and correlated inversely with lung function and body mass index. Follistatin treatment of newborn β-ENaC mice, noted for respiratory pathology mimicking human CF, decreased the airway activin A levels and key features of CF lung disease including mucus hypersecretion, airway neutrophilia and levels of mediators that regulate inflammation and chemotaxis. Follistatin treatment also increased body weight and survival of β-ENaC mice, with no evidence of local or systemic toxicity. Our findings demonstrate that activin A levels are elevated in CF and provide proof-of-concept for the use of the activin A antagonist, follistatin, as a therapeutic in the long-term management of lung disease in CF patients.
- Published
- 2015
- Full Text
- View/download PDF
30. Development of novel activin-targeted therapeutics.
- Author
-
Chen JL, Walton KL, Al-Musawi SL, Kelly EK, Qian H, La M, Lu L, Lovrecz G, Ziemann M, Lazarus R, El-Osta A, Gregorevic P, and Harrison CA
- Subjects
- Activins antagonists & inhibitors, Activins genetics, Animals, Bone Morphogenetic Proteins genetics, Bone Morphogenetic Proteins metabolism, Dependovirus genetics, Gene Expression Regulation, Genetic Vectors genetics, Growth Differentiation Factors genetics, Growth Differentiation Factors metabolism, HEK293 Cells, Humans, Immunoglobulin Fc Fragments genetics, Immunoglobulin Fc Fragments metabolism, Immunoglobulin G genetics, Immunoglobulin G metabolism, Male, Mice, Mice, Inbred C57BL, Muscle, Skeletal pathology, Myostatin genetics, Myostatin metabolism, Plasmids chemistry, Plasmids metabolism, Protein Structure, Tertiary, Recombinant Fusion Proteins genetics, Transfection, Transforming Growth Factor beta genetics, Transforming Growth Factor beta metabolism, Activins metabolism, Genetic Engineering methods, Muscle, Skeletal metabolism, Recombinant Fusion Proteins metabolism, Signal Transduction
- Abstract
Soluble activin type II receptors (ActRIIA/ActRIIB), via binding to diverse TGF-β proteins, can increase muscle and bone mass, correct anemia or protect against diet-induced obesity. While exciting, these multiple actions of soluble ActRIIA/IIB limit their therapeutic potential and highlight the need for new reagents that target specific ActRIIA/IIB ligands. Here, we modified the activin A and activin B prodomains, regions required for mature growth factor synthesis, to generate specific activin antagonists. Initially, the prodomains were fused to the Fc region of mouse IgG2A antibody and, subsequently, "fastener" residues (Lys(45), Tyr(96), His(97), and Ala(98); activin A numbering) that confer latency to other TGF-β proteins were incorporated. For the activin A prodomain, these modifications generated a reagent that potently (IC(50) 5 nmol/l) and specifically inhibited activin A signaling in vitro, and activin A-induced muscle wasting in vivo. Interestingly, the modified activin B prodomain inhibited both activin A and B signaling in vitro (IC(50) ~2 nmol/l) and in vivo, suggesting it could serve as a general activin antagonist. Importantly, unlike soluble ActRIIA/IIB, the modified prodomains did not inhibit myostatin or GDF-11 activity. To underscore the therapeutic utility of specifically antagonising activin signaling, we demonstrate that the modified activin prodomains promote significant increases in muscle mass.
- Published
- 2015
- Full Text
- View/download PDF
31. Over-expression of Follistatin-like 3 attenuates fat accumulation and improves insulin sensitivity in mice.
- Author
-
Brandt C, Hansen RH, Hansen JB, Olsen CH, Galle P, Mandrup-Poulsen T, Gehl J, Pedersen BK, and Hojman P
- Subjects
- Activins antagonists & inhibitors, Activins metabolism, Animals, Diet, High-Fat adverse effects, Female, Follistatin-Related Proteins, Glucagon metabolism, Glucose Intolerance etiology, Glucose Intolerance metabolism, Glucose Intolerance pathology, Glucose Intolerance prevention & control, Insulin metabolism, Islets of Langerhans cytology, Islets of Langerhans growth & development, Islets of Langerhans metabolism, Islets of Langerhans pathology, Liver growth & development, Liver metabolism, Liver pathology, Mice, Inbred C57BL, Muscle Development, Muscle, Skeletal growth & development, Muscle, Skeletal pathology, Myostatin antagonists & inhibitors, Myostatin metabolism, Proteins genetics, Random Allocation, Recombinant Proteins metabolism, Subcutaneous Fat growth & development, Subcutaneous Fat pathology, Adiposity, Insulin Resistance, Muscle, Skeletal metabolism, Proteins metabolism, Signal Transduction, Subcutaneous Fat metabolism, Up-Regulation
- Abstract
Objective: Follistatin-like 3 (fstl3), a natural inhibitor of members of the TGF-β family, increases during resistance training in human plasma. Fstl3 primarily binds myostatin and activin A, and thereby inhibits their functions. We hypothesize that blocking myostatin and activin A signalling through systemic fstl3 over-expression protects against diet-induced obesity and insulin resistance., Methods: Fstl3 was over-expressed by DNA electrotransfer in tibialis anterior, quadriceps and gastrocnemius muscles in female C57BL/C mice, and the mice were subsequently randomized to chow or high-fat feeding. Body weight, food intake, fat accumulation by MR scanning, and glucose, insulin and glucagon tolerance were evaluated, as was the response in body weight and metabolic parameters to 24h fasting. Effects of fstl3 on pancreatic insulin and glucagon content, and pancreatic islet morphology were determined., Results: Fstl3 over-expression reduced fat accumulation during high-fat feeding by 16%, and liver fat by 50%, as determined by MRI. No changes in body weight were observed, while the weight of the transfected muscles increased by 10%. No transcriptional changes were found in the subcutaneous adipose tissue. Fstl3 mice displayed improved insulin sensitivity and muscle insulin signalling. In contrast, glucose tolerance was impaired in high-fat fed fstl3 mice, which was explained by increased hepatic glucagon sensitivity and glucose output, as well as a decrease in the pancreatic insulin/glucagon ratio. Accordingly, fstl3 transfection improved counter-regulation to 24h fasting., Conclusion: Fstl3 over-expression regulates insulin and glucagon sensitivities through increased muscular insulin action, as well as increased hepatic glucagon sensitivity and pancreatic glucagon content., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
32. Experimental and investigational therapies for chemotherapy-induced anemia.
- Author
-
Testa U, Castelli G, and Elvira P
- Subjects
- Activins antagonists & inhibitors, Anemia chemically induced, Anemia physiopathology, Animals, Antineoplastic Agents therapeutic use, Disease Progression, Drugs, Investigational adverse effects, Drugs, Investigational pharmacology, Drugs, Investigational therapeutic use, Hematinics adverse effects, Hematinics pharmacology, Hematinics therapeutic use, Hepcidins antagonists & inhibitors, Humans, Neoplasms drug therapy, Anemia drug therapy, Antineoplastic Agents adverse effects, Drug Design
- Abstract
Introduction: In cancer patients, anemia is frequently observed, particularly as a consequence to chemotherapy (chemotherapy-induced anemia, CIA). CIA is treated with Red Blood Cell transfusions and erythropoiesis-stimulating agents (ESAs). However, the use of ESAs in anemic cancer patients is associated with reduced survival time and time to progression. Consequently, new therapeutic options are needed., Areas Covered: In this article, the authors discuss new erythroid-enhancing agents (EEAs) that act differently to erythropoietin. Specifically, the article summarizes the early clinical development of activin antagonists (Sotatercep [ACE-011] and ACE-536) and hepcidin antagonists [NOX-H94])., Expert Opinion: Both Activin RIIA trap agents and hepcidin inhibitors are promising new EEAs, but their safety profile, and their impact on treating CIA, needs to be carefully assessed in controlled clinical trials over longer periods of time. It is also important to carefully evaluate CIA patients to properly assess the physiopathological mechanisms responsible for the development of their anemic condition and provide patients with the most appropriate treatment plan.
- Published
- 2015
- Full Text
- View/download PDF
33. Sex specific retinoic acid signaling is required for the initiation of urogenital sinus bud development.
- Author
-
Bryant SL, Francis JC, Lokody IB, Wang H, Risbridger GP, Loveland KL, and Swain A
- Subjects
- Activins antagonists & inhibitors, Animals, DNA Primers, Female, Immunohistochemistry, In Situ Hybridization, Inhibin-beta Subunits antagonists & inhibitors, Male, Mice, Prostate embryology, Real-Time Polymerase Chain Reaction, Sex Factors, Tretinoin pharmacology, Urogenital System metabolism, beta-Galactosidase, Activins metabolism, Organogenesis physiology, Signal Transduction physiology, Tretinoin metabolism, Urogenital System embryology
- Abstract
The mammalian urogenital sinus (UGS) develops in a sex specific manner, giving rise to the prostate in the male and the sinus vagina in the embryonic female. Androgens, produced by the embryonic testis, have been shown to be crucial to this process. In this study we show that retinoic acid signaling is required for the initial stages of bud development from the male UGS. Enzymes involved in retinoic acid synthesis are expressed in the UGS mesenchyme in a sex specific manner and addition of ligand to female tissue is able to induce prostate-like bud formation in the absence of androgens, albeit at reduced potency. Functional studies in mouse organ cultures that faithfully reproduce the initiation of prostate development indicate that one of the roles of retinoic acid signaling in the male is to inhibit the expression of Inhba, which encodes the βA subunit of Activin, in the UGS mesenchyme. Through in vivo genetic analysis and culture studies we show that inhibition of Activin signaling in the female UGS leads to a similar phenotype to that of retinoic acid treatment, namely bud formation in the absence of androgens. Our data also reveals that both androgens and retinoic acid have extra independent roles to that of repressing Activin signaling in the development of the prostate during fetal stages. This study identifies a novel role for retinoic acid as a mesenchymal factor that acts together with androgens to determine the position and initiation of bud development in the male UGS epithelia., (Copyright © 2014 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
34. Conversion of human pancreatic acinar cells toward a ductal-mesenchymal phenotype and the role of transforming growth factor β and activin signaling.
- Author
-
De Waele E, Wauters E, Ling Z, and Bouwens L
- Subjects
- Acinar Cells cytology, Acinar Cells metabolism, Activins antagonists & inhibitors, Biomarkers, Bone Morphogenetic Protein 4 antagonists & inhibitors, Bone Morphogenetic Protein 4 pharmacology, Cell Lineage, Cells, Cultured, Humans, Plant Lectins, Receptors, Transforming Growth Factor beta antagonists & inhibitors, SOX9 Transcription Factor biosynthesis, SOX9 Transcription Factor genetics, Signal Transduction drug effects, Snail Family Transcription Factors, Suspensions, TGF-beta Superfamily Proteins antagonists & inhibitors, Transcription Factors biosynthesis, Transcription Factors genetics, Vimentin biosynthesis, Vimentin genetics, Acinar Cells drug effects, Activins physiology, Benzamides pharmacology, Cell Culture Techniques, Dioxoles pharmacology, Epithelial-Mesenchymal Transition drug effects, Pancreas, Exocrine cytology, TGF-beta Superfamily Proteins physiology
- Abstract
Objective: Epithelial-mesenchymal transition may interfere with the differentiation of cultured pancreatic acinar cells toward endocrine cells. Therefore, it will be important to investigate into detail the reprogramming of human pancreatic acinar cells toward a mesenchymal phenotype: the association with acinoductal transdifferentiation, the influence of cell adhesion, and the regulation behind this process., Methods: Human exocrine cells, isolated from donor pancreata, were cultured in suspension or as monolayers. Non-genetic lineage tracing, using labeled ulex europaeus agglutinin 1 lectin, was performed, and the role of the transforming growth factor (TGF-β) superfamily was investigated., Results: After 7 days in monolayer culture, the human acinar cells coexpressed the mesenchymal marker vimentin and the ductal marker Sox9. However, when the human exocrine cells were cultured in suspension, epithelial-mesenchymal transition was not observed. The spontaneous transition of the human acinar cells toward a ductal and mesenchymal phenotype was decreased by inhibition of the TGF-β and activin signaling pathways., Conclusions: The human acinar cells spontaneously undergo TGF-β- regulated reprogramming in the monolayer culture. These observations are helpful to develop culture methods for the in vitro reprogramming of pancreatic exocrine to endocrine cells. They are also of potential interest for studies on exocrine acinar cells in the development of pancreatic cancer.
- Published
- 2014
- Full Text
- View/download PDF
35. Ulipristal acetate modulates the expression and functions of activin a in leiomyoma cells.
- Author
-
Ciarmela P, Carrarelli P, Islam MS, Janjusevic M, Zupi E, Tosti C, Castellucci M, and Petraglia F
- Subjects
- Activins physiology, Adult, Female, Humans, Leiomyoma drug therapy, Middle Aged, Norpregnadienes therapeutic use, Tumor Cells, Cultured, Uterine Neoplasms drug therapy, Activins antagonists & inhibitors, Activins biosynthesis, Gene Expression Regulation, Neoplastic, Leiomyoma metabolism, Norpregnadienes pharmacology, Uterine Neoplasms metabolism
- Abstract
Uterine leiomyoma is the most common benign gynecological tumor in women of reproductive age and represents the single most common indication for hysterectomy. A development of new treatments is necessary for a medical management, and in this direction, several hormonal drugs are under investigation. Ulipristal acetate (UPA; a selective progesterone receptor modulator) is considered as one of the most promising because progesterone has a critical role in development and growth of uterine leiomyoma. The effect of steroids is partly mediated by growth factors like activin A which increases extracellular matrix expression contributing to the growth of leiomyoma. The present study aimed to test whether UPA acts on leiomyoma cells affecting expression and functions of activin A system. Cultured myometrial and leiomyoma cells were treated with UPA, and messenger RNA (mRNA) expression levels of activin A (inhibin βA [INHBA] subunits), its binding proteins (follistatin [FST] and FST-related gene), and its receptors (activin receptor-like kinase 4 [ALK4], activin receptor type [ActR] II, and ActRIIB) were evaluated. The effect of UPA on activin A modulation of fibronectin and vascular endothelial growth factor A (VEGF-A) mRNA expression in cultured myometrial and leiomyoma cells was also studied. Ulipristal acetate decreased INHBA, FST, ActRIIB, and Alk4 mRNA expressions in leiomyoma cultured cells. In addition, UPA was able to block the activin A-induced increase in fibronectin or VEGF-A mRNA expression in myometrial and in leiomyoma cultured cells. The present data show that UPA inhibits activin A expression and functions in leiomyoma cells, and this may represent a possible mechanism of action of the drug on uterine leiomyoma., (© The Author(s) 2014.)
- Published
- 2014
- Full Text
- View/download PDF
36. Bone morphogenetic protein 2 stimulates noncanonical SMAD2/3 signaling via the BMP type 1A receptor in gonadotrope-like cells: implications for FSH synthesis.
- Author
-
Wang Y, Ho CC, Bang E, Rejon CA, Libasci V, Pertchenko P, Hébert TE, and Bernard DJ
- Subjects
- Activins antagonists & inhibitors, Activins metabolism, Animals, Bone Morphogenetic Protein 2 agonists, Bone Morphogenetic Protein 2 antagonists & inhibitors, Bone Morphogenetic Protein 2 genetics, Bone Morphogenetic Protein Receptors, Type I agonists, Bone Morphogenetic Protein Receptors, Type I antagonists & inhibitors, Bone Morphogenetic Protein Receptors, Type I genetics, Cell Line, Follicle Stimulating Hormone, beta Subunit genetics, Follicle Stimulating Hormone, beta Subunit metabolism, Gene Silencing, Genes, Reporter, Humans, Mice, Phosphorylation, Protein Processing, Post-Translational, RNA, Small Interfering, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Smad2 Protein antagonists & inhibitors, Smad2 Protein genetics, Smad3 Protein antagonists & inhibitors, Smad3 Protein genetics, Transcription, Genetic, Bone Morphogenetic Protein 2 metabolism, Bone Morphogenetic Protein Receptors, Type I metabolism, Follicle Stimulating Hormone, beta Subunit biosynthesis, Gonadotrophs metabolism, Signal Transduction, Smad2 Protein metabolism, Smad3 Protein metabolism
- Abstract
FSH is an essential regulator of mammalian reproduction. Its synthesis by pituitary gonadotrope cells is regulated by multiple endocrine and paracrine factors, including TGFβ superfamily ligands, such as the activins and inhibins. Activins stimulate FSH synthesis via transcriptional regulation of its β-subunit gene (Fshb). More recently, bone morphogenetic proteins (BMPs) were shown to stimulate murine Fshb transcription alone and in synergy with activins. BMP2 signals via its canonical type I receptor, BMPR1A (or activin receptor-like kinase 3 [ALK3]), and SMAD1 and SMAD5 to stimulate transcription of inhibitor of DNA binding proteins. Inhibitor of DNA binding proteins then potentiate the actions of activin-stimulated SMAD3 to regulate the Fshb gene in the gonadotrope-like LβT2 cell line. Here, we report the unexpected observation that BMP2 also stimulates the SMAD2/3 pathway in these cells and that it does so directly via ALK3. Indeed, this novel, noncanonical ALK3 activity is completely independent of ALK4, ALK5, and ALK7, the type I receptors most often associated with SMAD2/3 pathway activation. Induction of the SMAD2/3 pathway by ALK3 is dependent upon its own previous activation by associated type II receptors, which phosphorylate conserved serine and threonine residues in the ALK3 juxtamembrane glycine-serine-rich domain. ALK3 signaling via SMAD3 is necessary for the receptor to stimulate Fshb transcription, whereas its activation of the SMAD1/5/8 pathway alone is insufficient. These data challenge current dogma that ALK3 and other BMP type I receptors signal via SMAD1, SMAD5, and SMAD8 and not SMAD2 or SMAD3. Moreover, they suggest that BMPs and activins may use similar intracellular signaling mechanisms to activate the murine Fshb promoter in immortalized gonadotrope-like cells.
- Published
- 2014
- Full Text
- View/download PDF
37. Inhibition of activin A ameliorates skeletal muscle injury and rescues contractile properties by inducing efficient remodeling in female mice.
- Author
-
Yaden BC, Wang YX, Wilson JM, Culver AE, Milner A, Datta-Mannan A, Shetler P, Croy JE, Dai G, and Krishnan V
- Subjects
- Animals, Electroporation, Enzyme-Linked Immunosorbent Assay, Female, Immunohistochemistry, Mice, Inbred C57BL, Regeneration physiology, Transcriptome, Activins antagonists & inhibitors, Muscle Contraction physiology, Muscle, Skeletal injuries, Muscle, Skeletal metabolism
- Abstract
Activin A, a member of the transforming growth factor-β superfamily, provides pleiotropic regulation of fibrosis and inflammation. We aimed at determining whether selective inhibition of activin A would provide a regenerative benefit. The introduction of activin A into normal muscle increased the expression of inflammatory and muscle atrophy genes Tnf, Tnfrsf12a, Trim63, and Fbxo32 by 3.5-, 10-, 2-, and 4-fold, respectively. The data indicate a sensitive response of muscle to activin A. Two hours after cardiotoxin-induced muscle damage, local activin A protein expression increased by threefold to ninefold. Neutralization of activin A with a specific monoclonal antibody in this muscle injury model decreased the muscle protein levels of lymphotoxin α and Il17a by 32% and 42%, respectively. Muscle histopathological features showed that activin A antibody-treated mice displayed an increase in muscle degradation, with the concomitant 9.2-fold elevation in F4/80-positive cells 3 days after injury. At the same time, the number of Pax7/Myod1-positive cells also increased, indicative of potentiated muscle precursor activation. Ultimately, activin A inhibition resulted in rapid recovery of muscle contractile properties indicated by a restoration of maximum and specific force. In summary, selective inhibition of activin A with a monoclonal antibody in muscle injury leads to the early onset of tissue degradation and subsequent enhanced myogenesis, thereby accelerating muscle repair and functional recovery., (Copyright © 2014 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
38. Activin A induces growth arrest through a SMAD- dependent pathway in hepatic progenitor cells.
- Author
-
Chen L, Zhang W, Liang HF, Zhou QF, Ding ZY, Yang HQ, Liu WB, Wu YH, Man Q, Zhang BX, and Chen XP
- Subjects
- Activins antagonists & inhibitors, Activins genetics, Animals, Cell Line, Cyclin D1 metabolism, Cyclin E metabolism, Cyclin-Dependent Kinase Inhibitor p15 genetics, Cyclin-Dependent Kinase Inhibitor p15 metabolism, Cyclin-Dependent Kinase Inhibitor p21 genetics, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Follistatin pharmacology, Hepatocytes physiology, Rats, Signal Transduction, Stem Cells physiology, Activins metabolism, Cell Proliferation, Hepatocytes metabolism, Smad Proteins metabolism, Stem Cells metabolism
- Abstract
Background: Activin A, an important member of transforming growth factor-β superfamily, is reported to inhibit proliferation of mature hepatocyte. However, the effect of activin A on growth of hepatic progenitor cells is not fully understood. To that end, we attempted to evaluate the potential role of activin A in the regulation of hepatic progenitor cell proliferation., Results: Using the 2-acetaminofluorene/partial hepatectomy model, activin A expression decreased immediately after partial hepatectomy and then increased from the 9th to 15th day post surgery, which is associated with the attenuation of oval cell proliferation. Activin A inhibited oval cell line LE6 growth via activating the SMAD signaling pathway, which manifested as the phosphorylation of SMAD2/3, the inhibition of Rb phosphorylation, the suppression of cyclinD1 and cyclinE, and the promotion of p21WAF1/Cip1 and p15INK4B expression. Treatment with activin A antagonist follistatin or blocking SMAD signaling could diminish the anti-proliferative effect of activin A. By contrast, inhibition of the MAPK pathway did not contribute to this effect. Antagonizing activin A activity by follistatin administration enhanced oval cell proliferation in the 2-acetylaminofluorene/partial hepatectomy model., Conclusion: Activin A, acting through the SMAD pathway, negatively regulates the proliferation of hepatic progenitor cells.
- Published
- 2014
- Full Text
- View/download PDF
39. Angiomodulin is required for cardiogenesis of embryonic stem cells and is maintained by a feedback loop network of p63 and Activin-A.
- Author
-
Wolchinsky Z, Shivtiel S, Kouwenhoven EN, Putin D, Sprecher E, Zhou H, Rouleau M, and Aberdam D
- Subjects
- Activins antagonists & inhibitors, Activins genetics, Animals, Cell Line, Embryonic Stem Cells metabolism, HeLa Cells, Humans, Inhibin-beta Subunits genetics, Inhibin-beta Subunits metabolism, Mice, Myocardium cytology, Neoplasm Proteins genetics, Phosphoproteins antagonists & inhibitors, Protein Binding, Protein Isoforms genetics, Protein Isoforms metabolism, RNA, Small Interfering metabolism, Recombinant Proteins genetics, Recombinant Proteins pharmacology, Signal Transduction, Trans-Activators antagonists & inhibitors, Activins metabolism, Cell Differentiation drug effects, Embryonic Stem Cells cytology, Feedback, Physiological drug effects, Neoplasm Proteins metabolism, Phosphoproteins metabolism, Trans-Activators metabolism
- Abstract
The transcription factor p63, member of the p53 gene family, encodes for two main isoforms, TAp63 and ΔNp63 with distinct functions on epithelial homeostasis and cancer. Recently, we discovered that TAp63 is essential for in vitro cardiogenesis and heart development in vivo. TAp63 is expressed by embryonic endoderm and acts on cardiac progenitors by a cell-non-autonomous manner. In the present study, we search for cardiogenic secreted factors that could be regulated by TAp63 and, by ChIP-seq analysis, identified Angiomodulin (AGM), also named IGFBP7 or IGFBP-rP1. We demonstrate that AGM is necessary for cardiac commitment of embryonic stem cells (ESCs) and its regulation depends on TAp63 isoform. TAp63 directly activates both AGM and Activin-A during ESC cardiogenesis while these secreted factors modulate TAp63 gene expression by a feedback loop mechanism. The molecular circuitry controlled by TAp63 on AGM/Activin-A signaling pathway and thus on cardiogenesis emphasizes the importance of p63 during early cardiac development., (© 2013.)
- Published
- 2014
- Full Text
- View/download PDF
40. Myostatin/activin pathway antagonism: molecular basis and therapeutic potential.
- Author
-
Han HQ, Zhou X, Mitch WE, and Goldberg AL
- Subjects
- Activins metabolism, Animals, Humans, Molecular Targeted Therapy, Muscle Proteins antagonists & inhibitors, Muscle Proteins metabolism, Muscle, Skeletal metabolism, Muscle, Skeletal pathology, Muscular Diseases metabolism, Muscular Diseases pathology, Myostatin metabolism, Proteasome Endopeptidase Complex metabolism, Signal Transduction, Ubiquitin metabolism, Ubiquitin-Protein Ligases metabolism, Activins antagonists & inhibitors, Muscular Diseases drug therapy, Myostatin antagonists & inhibitors
- Abstract
Muscle wasting is associated with a wide range of catabolic diseases. This debilitating loss of muscle mass and functional capacity reduces the quality of life and increases the risks of morbidity and mortality. Major progress has been made in understanding the biochemical mechanisms and signaling pathways regulating muscle protein balance under normal conditions and the enhanced protein loss in atrophying muscles. It is now clear that activation of myostatin/activin signaling is critical in triggering the accelerated muscle catabolism that causes muscle loss in multiple disease states. Binding of myostatin and activin to the ActRIIB receptor complex on muscle cell membrane leads to activation of Smad2/3-mediated transcription, which in turn stimulates FoxO-dependent transcription and enhanced muscle protein breakdown via ubiquitin-proteasome system and autophagy. In addition, Smad activation inhibits muscle protein synthesis by suppressing Akt signaling. Pharmacological blockade of the myostatin/activin-ActRIIB pathway has been shown to prevent or reverse the loss of muscle mass and strength in various disease models including cancer cachexia and renal failure. Moreover, it can markedly prolong the lifespan of animals with cancer-associated muscle loss. Furthermore, inhibiting myostatin/activin actions also improves insulin sensitivity, reduces excessive adiposity, attenuates systemic inflammation, and accelerates bone fracture healing in disease models. Based on these exciting advances, the potential therapeutic benefits of myostatin/activin antagonism are now being tested in multiple clinical settings. This article is part of a Directed Issue entitled: Molecular basis of muscle wasting., (Copyright © 2013. Published by Elsevier Ltd.)
- Published
- 2013
- Full Text
- View/download PDF
41. The therapeutic potential of blocking the activin signalling pathway.
- Author
-
Marino FE, Risbridger G, and Gold E
- Subjects
- Activins genetics, Activins metabolism, Animals, Cachexia genetics, Cachexia metabolism, Cell Proliferation drug effects, Humans, Musculoskeletal Diseases genetics, Musculoskeletal Diseases metabolism, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Neoplasms, Gonadal Tissue genetics, Neoplasms, Gonadal Tissue metabolism, Neuromuscular Diseases genetics, Neuromuscular Diseases metabolism, Signal Transduction genetics, Transforming Growth Factor beta genetics, Transforming Growth Factor beta metabolism, Activins antagonists & inhibitors, Antineoplastic Agents therapeutic use, Cachexia drug therapy, Musculoskeletal Diseases drug therapy, Neoplasm Proteins antagonists & inhibitors, Neoplasms, Gonadal Tissue drug therapy, Neuromuscular Diseases drug therapy, Signal Transduction drug effects
- Abstract
Members of the transforming growth factor β (TGF-β) family regulate fundamental physiological process, such as cell growth, differentiation and apoptosis. As a result, defects in this pathway have been linked to uncontrolled proliferation and cancer progression. Here we explore the signal transduction mechanism of TGF-β focusing on therapeutic intervention in human diseases. Like TGF-β, another member of the TGF-β superfamily, activin has been proven to play an important role in maintenance of tissue homeostasis and dysregulation leads to disease. Several studies showed elevated levels of activin are responsible for the development of gonadal tumours and a cachexia-like weight loss syndrome. Discussing the recent advances in approaches developed to antagonise the activin pathway and the encouraging results obtained in animal models, this review presents a therapeutic rationale for targeting the activin pathway in conditions such as cachexia, neuromuscular and/or musculoskeletal disorders., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
42. Development of a small-molecule screening method for inhibitors of cellular response to myostatin and activin A.
- Author
-
Cash JN, Angerman EB, Kirby RJ, Merck L, Seibel WL, Wortman MD, Papoian R, Nelson S, and Thompson TB
- Subjects
- Computer Simulation, Gene Expression drug effects, Genes, Reporter, HEK293 Cells, High-Throughput Screening Assays, Humans, Luciferases, Renilla biosynthesis, Luciferases, Renilla genetics, Small Molecule Libraries, Activins antagonists & inhibitors, Growth Substances pharmacology, Myostatin antagonists & inhibitors
- Abstract
Myostatin, a member of the transforming growth factor (TGF)-β family of secreted ligands, is a strong negative regulator of muscle growth. As such, therapeutic inhibitors of myostatin are actively being investigated for their potential in the treatment of muscle-wasting diseases such as muscular dystrophy and sarcopenia. Here, we sought to develop a high-throughput screening (HTS) method for small-molecule inhibitors that target myostatin. We created a HEK293 stable cell line that expresses the (CAGA)12-luciferase reporter construct and robustly responds to signaling of certain classes of TGF-β family ligands. After optimization and miniaturization of the assay to a 384-well format, we successfully screened a library of compounds for inhibition of myostatin and the closely related activin A. Selection of some of the tested compounds was directed by in silico screening against myostatin, which led to an enrichment of target hits as compared with random selection. Altogether, we present an HTS method that will be useful for screening potential inhibitors of not only myostatin but also many other ligands of the TGF-β family.
- Published
- 2013
- Full Text
- View/download PDF
43. Lenalidomide in combination with an activin A-neutralizing antibody: preclinical rationale for a novel anti-myeloma strategy.
- Author
-
Scullen T, Santo L, Vallet S, Fulciniti M, Eda H, Cirstea D, Patel K, Nemani N, Yee A, Mahindra A, and Raje N
- Subjects
- Activins metabolism, Cell Differentiation drug effects, Cell Line, Tumor, Humans, Lenalidomide, MAP Kinase Signaling System drug effects, Multiple Myeloma genetics, Osteoblasts cytology, Osteoblasts drug effects, Osteoblasts metabolism, Stromal Cells drug effects, Stromal Cells metabolism, Thalidomide pharmacology, Activins antagonists & inhibitors, Angiogenesis Inhibitors pharmacology, Antibodies, Neutralizing pharmacology, Antineoplastic Agents pharmacology, Multiple Myeloma metabolism, Thalidomide analogs & derivatives
- Abstract
Given the prevalence of osteolytic bone disease in multiple myeloma (MM), novel therapies targeting bone microenvironment are essential. Previous studies have identified activin A to be of critical importance in MM-induced osteolysis. Lenalidomide is a known and approved treatment strategy for relapsed MM. Our findings demonstrate that lenalidomide acts directly on bone marrow stromal cells via an Akt-mediated increase in Jun N-terminal kinase-dependent signaling resulting in activin A secretion, with consequent inhibition of osteoblastogenesis. Here, we attempted to augment the antitumor benefits of lenalidomide while overcoming its effects on osteoblastogenesis by combining it with a neutralizing antibody to activin A. Increased activin A secretion induced by lenalidomide was abrogated by the addition of activin A-neutralizing antibody, which effectively restored osteoblast function and inhibited MM-induced osteolysis without negating the cytotoxic effects of lenalidomide on malignant cells. This provides the rationale for an ongoing clinical trial (NCT01562405) combining lenalidomide with an anti-activin A strategy.
- Published
- 2013
- Full Text
- View/download PDF
44. Exercise restores decreased physical activity levels and increases markers of autophagy and oxidative capacity in myostatin/activin-blocked mdx mice.
- Author
-
Hulmi JJ, Oliveira BM, Silvennoinen M, Hoogaars WM, Pasternack A, Kainulainen H, and Ritvos O
- Subjects
- Activin Receptors, Type II biosynthesis, Activins physiology, Adiposity genetics, Adiposity physiology, Animals, Blotting, Western, Body Weight physiology, Citrate (si)-Synthase metabolism, Creatine Kinase blood, DNA biosynthesis, DNA isolation & purification, Eating physiology, Hematocrit, Hemoglobins metabolism, Mice, Mice, Inbred C57BL, Mice, Inbred mdx, Muscle, Skeletal metabolism, Muscle, Skeletal physiology, Myostatin physiology, Oxidation-Reduction, Tumor Necrosis Factor-alpha metabolism, Activin Receptors, Type II pharmacology, Activins antagonists & inhibitors, Autophagy physiology, Motor Activity physiology, Myostatin antagonists & inhibitors, Physical Conditioning, Animal physiology
- Abstract
The importance of adequate levels of muscle size and function and physical activity is widely recognized. Myostatin/activin blocking increases skeletal muscle mass but may decrease muscle oxidative capacity and can thus be hypothesized to affect voluntary physical activity. Soluble activin receptor IIB (sActRIIB-Fc) was produced to block myostatin/activins. Modestly dystrophic mdx mice were injected with sActRIIB-Fc or PBS with or without voluntary wheel running exercise for 7 wk. Healthy mice served as controls. Running for 7 wk attenuated the sActRIIB-Fc-induced increase in body mass by decreasing fat mass. Running also enhanced/restored the markers of muscle oxidative capacity and autophagy in mdx mice to or above the levels of healthy mice. Voluntary running activity was decreased by sActRIIB-Fc during the first 3-4 wk correlating with increased body mass. Home cage physical activity of mice, quantified from the force plate signal, was decreased by sActRIIB-Fc the whole 7-wk treatment in sedentary mice. To understand what happens during the first weeks after sActRIIB-Fc administration, when mice are less active, healthy mice were injected with sActRIIB-Fc or PBS for 2 wk. During the sActRIIB-Fc-induced rapid 2-wk muscle growth period, oxidative capacity and autophagy were reduced, which may possibly explain the decreased running activity. These results show that increased muscle size and decreased markers of oxidative capacity and autophagy during the first weeks of myostatin/activin blocking are associated with decreased voluntary activity levels. Voluntary exercise in dystrophic mice enhances the markers of oxidative capacity and autophagy to or above the levels of healthy mice.
- Published
- 2013
- Full Text
- View/download PDF
45. [Role of Activin A and Myostatin in cancer cachexia].
- Author
-
Thissen JP and Loumaye A
- Subjects
- Activins antagonists & inhibitors, Activins genetics, Activins pharmacology, Animals, Cachexia genetics, Cachexia metabolism, Cachexia prevention & control, Humans, Mice, Mice, Knockout, Myostatin antagonists & inhibitors, Myostatin metabolism, Myostatin pharmacology, Neoplasms drug therapy, Neoplasms genetics, Neoplasms pathology, Activins physiology, Cachexia etiology, Myostatin physiology, Neoplasms complications
- Abstract
Recent works suggest that Activin A (ActA) and Myostatin (Mstn), two members of the TGFβ superfamily, could contribute to skeletal muscle atrophy observed in some cancers. It is known that several human tumoral cell lines synthesize and secrete ActA and Mstn. In addition, systemic treatment with ActA and Mstn in mice induce muscle atrophy. Likewise, Inhibin-α knock-out mice, which are characterized by elevated circulating levels of ActA, exhibit muscle atrophy and die of cachexia. Finally, administration of ActA and Mstn antagonists prevents muscular atrophy and mortality induced by some animal tumors. Collectively, these findings suggest that ActA or Mstn production by several cancers could contribute to cachexia and thus to mortality associated with some cancers in human. This hypothesis is very interesting since new molecules that are able to inhibit ActA and Mstn, in particularly the sActRIIB, are under development., (Copyright © 2013 Elsevier Masson SAS. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
46. Multiple roles of Activin/Nodal, bone morphogenetic protein, fibroblast growth factor and Wnt/β-catenin signalling in the anterior neural patterning of adherent human embryonic stem cell cultures.
- Author
-
Lupo G, Novorol C, Smith JR, Vallier L, Miranda E, Alexander M, Biagioni S, Pedersen RA, and Harris WA
- Subjects
- Activins antagonists & inhibitors, Benzamides pharmacology, Bone Morphogenetic Proteins antagonists & inhibitors, Carrier Proteins pharmacology, Cells, Cultured, Dioxoles pharmacology, Embryonic Stem Cells cytology, Fibroblast Growth Factor 2 pharmacology, Gene Expression Regulation, Developmental drug effects, Hedgehog Proteins metabolism, Humans, Neural Plate metabolism, Nodal Protein antagonists & inhibitors, Signal Transduction drug effects, Up-Regulation drug effects, Wnt Proteins antagonists & inhibitors, Activins metabolism, Bone Morphogenetic Proteins metabolism, Embryonic Stem Cells metabolism, Fibroblast Growth Factors metabolism, Nodal Protein metabolism, Wnt Proteins metabolism, beta Catenin metabolism
- Abstract
Several studies have successfully produced a variety of neural cell types from human embryonic stem cells (hESCs), but there has been limited systematic analysis of how different regional identities are established using well-defined differentiation conditions. We have used adherent, chemically defined cultures to analyse the roles of Activin/Nodal, bone morphogenetic protein (BMP), fibroblast growth factor (FGF) and Wnt/β-catenin signalling in neural induction, anteroposterior patterning and eye field specification in hESCs. We show that either BMP inhibition or activation of FGF signalling is required for effective neural induction, but these two pathways have distinct outcomes on rostrocaudal patterning. While BMP inhibition leads to specification of forebrain/midbrain positional identities, FGF-dependent neural induction is associated with strong posteriorization towards hindbrain/spinal cord fates. We also demonstrate that Wnt/β-catenin signalling is activated during neural induction and promotes acquisition of neural fates posterior to forebrain. Therefore, inhibition of this pathway is needed for efficient forebrain specification. Finally, we provide evidence that the levels of Activin/Nodal and BMP signalling have a marked influence on further forebrain patterning and that constitutive inhibition of these pathways represses expression of eye field genes. These results show that the key mechanisms controlling neural patterning in model vertebrate species are preserved in adherent, chemically defined hESC cultures and reveal new insights into the signals regulating eye field specification.
- Published
- 2013
- Full Text
- View/download PDF
47. Activin A is essential for Feeder-free culture of human induced pluripotent stem cells.
- Author
-
Tomizawa M, Shinozaki F, Sugiyama T, Yamamoto S, Sueishi M, and Yoshida T
- Subjects
- Activins antagonists & inhibitors, Alkaline Phosphatase analysis, Animals, Antigens, Surface analysis, Benzamides pharmacology, Collagen, Dioxoles pharmacology, Drug Combinations, Fibroblast Growth Factor 2 pharmacology, Homeodomain Proteins analysis, Humans, Laminin, Leukemia Inhibitory Factor pharmacology, Mice, Nanog Homeobox Protein, Octamer Transcription Factor-3 analysis, Proteoglycans analysis, Stage-Specific Embryonic Antigens analysis, Activins pharmacology, Cell Culture Techniques, Feeder Cells, Induced Pluripotent Stem Cells metabolism, Pyridines pharmacology, Pyrimidines pharmacology
- Abstract
Feeder-free culture of human induced pluripotent stem (hiPS) cells is necessary for their clinical application to avoid adverse effects of foreign proteins. hiPS cells were cultured with combinations of activin (A), CHIR99021 (C), basic fibroblast growth factor (F), and leukemia inhibitory factor (L) under feeder-free conditions. Culture was terminated after 12 passages or when the cell morphology changed from pluripotency. Pluripotency was analyzed by alkaline phosphatase (ALP) staining and immunostaining with antibodies to Oct3/4, Nanog, SSEA4, and TRA-1-60. SB431542 (SB), an activin inhibitor, was added to the culture, and the morphology of the cells was observed. hiPS cells cultured with A, AC, and ACL after 12 passages were positive for ALP staining. Oct3/4 was positive in hiPS cells cultured with A, AC, and ACL. hiPS cells were positive for Nanog when cultured with A and AC; however, Nanog signal was weaker in cells cultured with ACL. SSEA4 was positive in hiPS cells cultured with A and AC but almost negative in those cultured with ACL. hiPS cells were positive for TRA-1-60 when cultured with A, AC, and ACL. hiPS cells lose their undifferentiated morphology at six passages when cultured with A + SB, five passages with AC + SB, and nine passages with ACL. We conclude that feeder-free culture of hiPS cells requires A or AC to maintain pluripotency., (Copyright © 2012 Wiley Periodicals, Inc.)
- Published
- 2013
- Full Text
- View/download PDF
48. Activins in adipogenesis and obesity.
- Author
-
Dani C
- Subjects
- Activins antagonists & inhibitors, Activins pharmacology, Adipogenesis, Animals, Biomarkers metabolism, Cell Differentiation, Cell Proliferation, Fibrosis therapy, Humans, Mice, Mice, Obese, Obesity prevention & control, Obesity therapy, Signal Transduction, Activins metabolism, Adipose Tissue metabolism, Fibrosis metabolism, Follistatin metabolism, Obesity metabolism
- Abstract
Activins are secreted proteins members of the transforming growth factor-β family. They are involved in many biological responses including regulation of apoptosis, proliferation and differentiation of different cell types. Activins A, B and AB are highly expressed in adipose tissue, and in this review we will illustrate that activins have a role in several steps of physiological and pathological development of adipose tissue. Activin A has been shown to be a critical regulator of human adipocyte progenitor proliferation and a potent inhibitor of their differentiation. Activin A could also be a mediator of fibrosis observed in obese adipose tissue. Activin B/AB is proposed as a new adipokine having a role in energy balance and insulin insensitivity associated with obesity. Therefore, activin pathway could represent a potential therapeutic target both for controlling the size and the phenotype of the adipose precursor pool and for obesity-associated metabolic complications.
- Published
- 2013
- Full Text
- View/download PDF
49. Follistatin antagonizes activin signaling and acts with notum to direct planarian head regeneration.
- Author
-
Roberts-Galbraith RH and Newmark PA
- Subjects
- Activins genetics, Activins physiology, Animals, Body Patterning genetics, Body Patterning physiology, Follistatin genetics, Gene Knockdown Techniques, Genes, Helminth, Helminth Proteins antagonists & inhibitors, Helminth Proteins genetics, Molecular Sequence Data, Planarians cytology, Planarians genetics, Pluripotent Stem Cells cytology, Pluripotent Stem Cells physiology, RNA Interference, Regeneration genetics, Regeneration physiology, Signal Transduction, Wnt Proteins antagonists & inhibitors, Activins antagonists & inhibitors, Follistatin physiology, Helminth Proteins physiology, Planarians physiology
- Abstract
Animals establish their body plans in embryogenesis, but only a few animals can recapitulate this signaling milieu for regeneration after injury. In planarians, a pluripotent stem cell population and perpetual signaling of polarity axes collaborate to direct a steady replacement of cells during homeostasis and to power robust regeneration after even severe injuries. Several studies have documented the roles of conserved signaling pathways in maintaining and resetting axial polarity in planarians, but it is unclear how planarians reestablish polarity signaling centers after injury and whether these centers serve to influence identity decisions of stem cell progeny during their differentiation. Here we find that a planarian Follistatin homolog directs regeneration of anterior identity by opposing an Activin/ActR-1/Smad2/3 signaling pathway. Follistatin and Notum, a Wnt inhibitor, are mutually required to reestablish an anterior signaling center that expresses both cues. Furthermore, we show that the direction of cells down particular differentiation paths requires regeneration of this anterior signaling center. Just as its amphibian counterpart in the organizer signals body plan and cell fate during embryogenesis, planarian Follistatin promotes reestablishment of anterior polarity during regeneration and influences specification of cell types in the head and beyond.
- Published
- 2013
- Full Text
- View/download PDF
50. The activin A antagonist follistatin inhibits asthmatic airway remodelling.
- Author
-
Hardy CL, Nguyen HA, Mohamud R, Yao J, Oh DY, Plebanski M, Loveland KL, Harrison CA, Rolland JM, and O'Hehir RE
- Subjects
- Administration, Intranasal, Airway Remodeling immunology, Analysis of Variance, Animals, Asthma immunology, Asthma pathology, Bronchoalveolar Lavage Fluid cytology, Bronchoalveolar Lavage Fluid immunology, Cytokines analysis, Disease Models, Animal, Enzyme-Linked Immunosorbent Assay, Female, Follistatin immunology, Immunohistochemistry, Interleukin-13 analysis, Interleukin-13 metabolism, Interleukin-4 analysis, Interleukin-4 metabolism, Interleukin-5 analysis, Interleukin-5 metabolism, Mice, Mice, Inbred BALB C, Ovalbumin immunology, Ovalbumin metabolism, Random Allocation, Reference Values, Sensitivity and Specificity, Transforming Growth Factor beta analysis, Activins antagonists & inhibitors, Airway Remodeling drug effects, Asthma drug therapy, Cytokines metabolism, Follistatin pharmacology, Transforming Growth Factor beta metabolism
- Abstract
Background: Current pharmacotherapy is highly effective in the clinical management of the majority of patients with stable asthma, however severe asthma remains inadequately treated. Prevention of airway remodelling is a major unmet clinical need in the management of patients with chronic severe asthma and other inflammatory lung diseases. Accumulating evidence convincingly demonstrates that activin A, a member of the transforming growth factor (TGF)-β superfamily, is a key driver of airway inflammation, but its role in chronic asthmatic airway remodelling is ill-defined. Follistatin, an endogenously produced protein, binds activin A with high affinity and inhibits its bioactivity. The aim of this study was to test the potential of follistatin as a therapeutic agent to inhibit airway remodelling in an experimental model of chronic allergic airway inflammation., Methods: BALB/c mice were systemically sensitised with ovalbumin (OVA), and challenged with OVA intranasally three times a week for 10 weeks. Follistatin was instilled intranasally during allergen challenge., Results: Chronic allergen challenge induced mucus hypersecretion and subepithelial collagen deposition which persisted after cessation of challenge. Intranasal follistatin (0.05, 0.5, 5 µg) inhibited the airway remodelling and dose-dependently decreased airway activin A and TGF-β1, and allergen-specific T helper 2 cytokine production in the lung-draining lymph nodes. Follistatin also impaired the loss of TGF-β1 and activin RIB immunostaining in airway epithelium which occurred following chronic allergen challenge., Conclusions: These data demonstrate that follistatin attenuates asthmatic airway remodelling. Our findings point to the potential of follistatin as a therapeutic for prevention of airway remodelling in asthma and other inflammatory lung diseases.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.