scholarly journals Functional redundancy of type I and type II receptors in the regulation of skeletal muscle growth by myostatin and activin A

2020 ◽  
Vol 117 (49) ◽  
pp. 30907-30917 ◽  
Author(s):  
Se-Jin Lee ◽  
Adam Lehar ◽  
Yewei Liu ◽  
Chi Hai Ly ◽  
Quynh-Mai Pham ◽  
...  

Myostatin (MSTN) is a transforming growth factor-β (TGF-β) family member that normally acts to limit muscle growth. The function of MSTN is partially redundant with that of another TGF-β family member, activin A. MSTN and activin A are capable of signaling through a complex of type II and type I receptors. Here, we investigated the roles of two type II receptors (ACVR2 and ACVR2B) and two type I receptors (ALK4 and ALK5) in the regulation of muscle mass by these ligands by genetically targeting these receptors either alone or in combination specifically in myofibers in mice. We show that targeting signaling in myofibers is sufficient to cause significant increases in muscle mass, showing that myofibers are the direct target for signaling by these ligands in the regulation of muscle growth. Moreover, we show that there is functional redundancy between the two type II receptors as well as between the two type I receptors and that all four type II/type I receptor combinations are utilized in vivo. Targeting signaling specifically in myofibers also led to reductions in overall body fat content and improved glucose metabolism in mice fed either regular chow or a high-fat diet, demonstrating that these metabolic effects are the result of enhanced muscling. We observed no effect, however, on either bone density or muscle regeneration in mice in which signaling was targeted in myofibers. The latter finding implies that MSTN likely signals to other cells, such as satellite cells, in addition to myofibers to regulate muscle homeostasis.

Blood ◽  
2008 ◽  
Vol 111 (5) ◽  
pp. 2733-2743 ◽  
Author(s):  
Neil C. Robson ◽  
David J. Phillips ◽  
Tristan McAlpine ◽  
Amanda Shin ◽  
Suzanne Svobodova ◽  
...  

Activin-A is a transforming growth factor-β (TGF-β) superfamily member that plays a pivotal role in many developmental and reproductive processes. It is also involved in neuroprotection, apoptosis of tumor and some immune cells, wound healing, and cancer. Its role as an immune-regulating protein has not previously been described. Here we demonstrate for the first time that activin-A has potent autocrine effects on the capacity of human dendritic cells (DCs) to stimulate immune responses. Human monocyte-derived DCs (MoDCs) and the CD1c+ and CD123+ peripheral blood DC populations express both activin-A and the type I and II activin receptors. Furthermore, MoDCs and CD1c+ myeloid DCs rapidly secrete high levels of activin-A after exposure to bacteria, specific toll-like receptor (TLR) ligands, or CD40 ligand (CD40L). Blocking autocrine activin-A signaling in DCs using its antagonist, follistatin, enhanced DC cytokine (IL-6, IL-10, IL-12p70, and tumor necrosis factor-α [TNF-α]) and chemokine (IL-8, IP-10, RANTES, and MCP-1) production during CD40L stimulation, but not TLR-4 ligation. Moreover, antagonizing DC-derived activin-A resulted in significantly enhanced expansion of viral antigen-specific effector CD8+ T cells. These findings establish an immune-regulatory role for activin-A in DCs, highlighting the potential of antagonizing activin-A signaling in vivo to enhance vaccine immunogenicity.


2009 ◽  
Vol 296 (6) ◽  
pp. C1248-C1257 ◽  
Author(s):  
Roberta Sartori ◽  
Giulia Milan ◽  
Maria Patron ◽  
Cristina Mammucari ◽  
Bert Blaauw ◽  
...  

Loss of muscle mass occurs in a variety of diseases, including cancer, chronic heart failure, aquired immunodeficiency syndrome, diabetes, and renal failure, often aggravating pathological progression. Preventing muscle wasting by promoting muscle growth has been proposed as a possible therapeutic approach. Myostatin is an important negative modulator of muscle growth during myogenesis, and myostatin inhibitors are attractive drug targets. However, the role of the myostatin pathway in adulthood and the transcription factors involved in the signaling are unclear. Moreover, recent results confirm that other transforming growth factor-β (TGF-β) members control muscle mass. Using genetic tools, we perturbed this pathway in adult myofibers, in vivo, to characterize the downstream targets and their ability to control muscle mass. Smad2 and Smad3 are the transcription factors downstream of myostatin/TGF-β and induce an atrophy program that is muscle RING-finger protein 1 (MuRF1) independent. Furthermore, Smad2/3 inhibition promotes muscle hypertrophy independent of satellite cells but partially dependent of mammalian target of rapamycin (mTOR) signaling. Thus myostatin and Akt pathways cross-talk at different levels. These findings point to myostatin inhibitors as good drugs to promote muscle growth during rehabilitation, especially when they are combined with IGF-1-Akt activators.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 261-261 ◽  
Author(s):  
Laura Silvestri ◽  
Silvia Colucci ◽  
Alessia Pagani ◽  
Irene Artuso ◽  
Mariatesera Pettinato ◽  
...  

Abstract Introduction Hepcidin, the liver hormone that regulates iron homeostasis, is mainly activated via the BMP/SMAD pathway. Among other stimuli, as erythropoiesis expansion, inflammation and hypoxia, hepcidin expression is influenced by drugs, including the mTOR inhibitor rapamycin (RAPA) (Mleczko-Sanecka et al., Blood 2014). In the liver, BMP type I receptors, ALK2 and ALK3, are both essential for hepcidin regulation (Steinbiecker et al., Blood 2012), whereas BMP type II receptors, BMPR2 and ACVR2A, have a redundant role (Mayeur et al., Blood 2014). The signaling is activated when BMP type II receptors phosphorylate residues of the glycine/serine rich (GS) domain of BMP type I receptors, which then activate SMAD1/5/8. Treatment with RAPA increases hepcidin expression in murine hepatocytes and may cause microcytic anemia in patients (Przybylowski P. et al., Transplant Proc. 2013). However, the mechanisms involved in hepcidin and mTOR crosstalk are unknown. Methods Hepcidin, BMP-SMAD and mTOR target genes were analyzed in human hepatoma cell lines and murine primary hepatocytes (HCs), treated with RAPA (100 nM), Torin1 (100 nM), tacrolimus (TAC, 1 μg/ml) in the presence or absence of the BMP pathway inhibitor DMH1 (0.5 μg/ml) or of the ligands BMP6, BMP2, Activin B (ActB) and Activin A (ActA) (1-100 ng/ml). ALK2wt was mutagenized in the GS domain (R206H, Q207E) or close to the GS domain (R258S) to generate ALK2mut with reduce binding to FKBP12 (Taylor et al., Cancer Res. 2014). SMAD1/5/8 phosphorylation was analyzed by Western Blotting in cells transfected with SMAD1 and ALK2wt or ALK2mut and treated or not with TAC, BMP6 and ActA. Binding of FKBP12 to ALK2wt and ALK2mut was assessed by coimmunoprecipitation of tagged proteins in transfected cells treated as above. Eight weeks old C57BL/6 wild type male mice were treated with a single dose of TAC (10 mg/kg) or vehicle and sacrificed at different time points (3-18 hrs). Hepcidin expression, LIC, SIC, serum iron and hematological parameters were analyzed by standard methods. Results We analyzed hepcidin expression in hepatoma cells and primary HCs treated with RAPA, an inhibitor of mTORC1, and Torin1, an ATP-competitive inhibitor of mTORC1 and 2. Hepcidin is increased by RAPA, but not Torin1, in a SMAD1/5/8 dependent way since DMH1 abrogates the effect. RAPA inhibits mTORC1 when complexed with FKBP12, an immunophilin that binds BMP receptors to avoid leakage activation of the pathway. To investigate whether hepcidin upregulation by RAPA is mediated by FKBP12 sequestration, we used genetic and pharmacologic approaches. First, we confirmed by coimmunoprecipitation that ALK2mut have a reduced ability to bind FKBP12. Then we transfected hepatoma cells with ALK2wt and ALK2mut and analyzed hepcidin and BMP pathway activation. Overexpression of ALK2mut increases hepcidin through SMAD1/5/8 as shown by high levels of SMAD1 phosphorylation, an effect abrogated by DMH1. Second, we treated hepatoma cells and primary HCs with TAC, a calcineurin inhibitor that binds FKBP12. This treatment increases hepcidin through SMAD1/5/8, suggesting a mechanism shared with RAPA. The same effect is observed in vivo since hepcidin is increased at 6 hrs post-injection in TAC-treated wt mice. Our results identify FKBP12 as a novel regulator of hepcidin. In addition, FKBP12 displacement alters the BMP receptor selectivity to ligands. Despite ALK2wt preferentially binds BMP6, ALK2mut become responsive to ActA, a TGF-β ligand that signals through SMAD2/3. Hepcidin activation by BMP6, BMP2 and ActB is comparable in ALK2wt and ALK2mut expressing cells. However, ActA upregulates hepcidin (through SMAD1/5/8) only in ALK2mut transfected cells. This effect is due to the impaired ability of ALK2mut to bind FKBP12, since it is observed even in ALK2wt transfected cells pretreated with TAC. Conclusions FKBP12 contributes to hepcidin regulation both in vitro and in vivo, thus adding a new player to the BMP-dependent hepcidin activation and a potential pharmacologic target for disorders characterized by low hepcidin and iron overload. Furthermore the ability of ALK2 to respond to Activin A, which is released in inflammation, links the BMP pathway-hepcidin activation to the inflammatory response. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Nozomi Igarashi ◽  
Megumi Honjo ◽  
Makoto Aihara

AbstractWe examined the effects of mTOR inhibitors on the fibrotic response induced by transforming growth factor-beta2 (TGF-β2) in cultured human trabecular meshwork (hTM) cells. TGF-β2-induced expression of fibronectin, collagen type I, alpha 1 chain (COL1A1), and alpha-smooth muscle actin (αSMA) in hTM cells was examined in the presence or absence of mTOR inhibitors using quantitative real-time polymerase chain reaction, Western blotting, and immunohistochemistry. The migration rates of hTM cells were examined in the presence of TGF-β2 with or without mTOR inhibitors. An in vitro study showed that the expression of fibronectin, COL1A1, and αSMA was upregulated by TGF-β2 treatment of hTM cells; such upregulation was significantly suppressed by mTOR inhibitors. The inhibitors significantly reduced the migration rate of TGF-β2-stimulated hTM cells. mTOR inhibitors may usefully reduce the fibrotic response of hTM cells and we may have to explore if it is also effective in in vivo model.


2001 ◽  
Vol 12 (3) ◽  
pp. 675-684 ◽  
Author(s):  
Jules J.E. Doré ◽  
Diying Yao ◽  
Maryanne Edens ◽  
Nandor Garamszegi ◽  
Elizabeth L. Sholl ◽  
...  

Transforming growth factor-βs (TGF-β) are multifunctional proteins capable of either stimulating or inhibiting mitosis, depending on the cell type. These diverse cellular responses are caused by stimulating a single receptor complex composed of type I and type II receptors. Using a chimeric receptor model where the granulocyte/monocyte colony-stimulating factor receptor ligand binding domains are fused to the transmembrane and cytoplasmic signaling domains of the TGF-β type I and II receptors, we wished to describe the role(s) of specific amino acid residues in regulating ligand-mediated endocytosis and signaling in fibroblasts and epithelial cells. Specific point mutations were introduced at Y182, T200, and Y249 of the type I receptor and K277 and P525 of the type II receptor. Mutation of either Y182 or Y249, residues within two putative consensus tyrosine-based internalization motifs, had no effect on endocytosis or signaling. This is in contrast to mutation of T200 to valine, which resulted in ablation of signaling in both cell types, while only abolishing receptor down-regulation in fibroblasts. Moreover, in the absence of ligand, both fibroblasts and epithelial cells constitutively internalize and recycle the TGF-β receptor complex back to the plasma membrane. The data indicate fundamental differences between mesenchymal and epithelial cells in endocytic sorting and suggest that ligand binding diverts heteromeric receptors from the default recycling pool to a pathway mediating receptor down-regulation and signaling.


2020 ◽  
Vol 117 (9) ◽  
pp. 4910-4920 ◽  
Author(s):  
Joonho Suh ◽  
Na-Kyung Kim ◽  
Seung-Hoon Lee ◽  
Je-Hyun Eom ◽  
Youngkyun Lee ◽  
...  

Growth and differentiation factor 11 (GDF11) and myostatin (MSTN) are closely related transforming growth factor β (TGF-β) family members, but their biological functions are quite distinct. While MSTN has been widely shown to inhibit muscle growth, GDF11 regulates skeletal patterning and organ development during embryogenesis. Postnatal functions of GDF11, however, remain less clear and controversial. Due to the perinatal lethality ofGdf11null mice, previous studies used recombinant GDF11 protein to prove its postnatal function. However, recombinant GDF11 and MSTN proteins share nearly identical biochemical properties, and most GDF11-binding molecules have also been shown to bind MSTN, generating the possibility that the effects mediated by recombinant GDF11 protein actually reproduce the endogenous functions of MSTN. To clarify the endogenous functions of GDF11, here, we focus on genetic studies and show thatGdf11null mice, despite significantly down-regulatingMstnexpression, exhibit reduced bone mass through impaired osteoblast (OB) and chondrocyte (CH) maturations and increased osteoclastogenesis, while the opposite is observed inMstnnull mice that display enhanced bone mass. Mechanistically,Mstndeletion up-regulatesGdf11expression, which activates bone morphogenetic protein (BMP) signaling pathway to enhance osteogenesis. Also, mice overexpressing follistatin (FST), a MSTN/GDF11 inhibitor, exhibit increased muscle mass accompanied by bone fractures, unlikeMstnnull mice that display increased muscle mass without fractures, indicating that inhibition of GDF11 impairs bone strength. Together, our findings suggest that GDF11 promotes osteogenesis in contrast to MSTN, and these opposing roles of GDF11 and MSTN must be considered to avoid the detrimental effect of GDF11 inhibition when developing MSTN/GDF11 inhibitors for therapeutic purposes.


2013 ◽  
Vol 126 (8) ◽  
pp. 557-566 ◽  
Author(s):  
Tim Snijders ◽  
Benjamin T. Wall ◽  
Marlou L. Dirks ◽  
Joan M. G. Senden ◽  
Fred Hartgens ◽  
...  

Two weeks of muscle disuse led to a loss in muscle mass and strength. The loss in muscle mass was attributed to both type I and type II muscle fibre atrophy, and was not accompanied by a decline in satellite cell content.


Sign in / Sign up

Export Citation Format

Share Document