Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters

Publication year range
1.
FASEB J ; 35(3): e21263, 2021 03.
Article in English | MEDLINE | ID: mdl-33570811

ABSTRACT

Bone is a dynamic tissue that constantly adapts to changing mechanical demands. The transforming growth factor beta (TGFß) signaling pathway plays several important roles in maintaining skeletal homeostasis by both coupling the bone-forming and bone-resorbing activities of osteoblasts and osteoclasts and by playing a causal role in the anabolic response of bone to applied loads. However, the extent to which the TGFß signaling pathway in osteocytes is directly regulated by fluid shear stress (FSS) is unknown, despite work suggesting that fluid flow along canaliculi is a dominant physical cue sensed by osteocytes following bone compression. To investigate the effects of FSS on TGFß signaling in osteocytes, we stimulated osteocytic OCY454 cells cultured within a microfluidic platform with FSS. We find that FSS rapidly upregulates Smad2/3 phosphorylation and TGFß target gene expression, even in the absence of added TGFß. Indeed, relative to treatment with TGFß, FSS induced a larger increase in levels of pSmad2/3 and Serpine1 that persisted even in the presence of a TGFß receptor type I inhibitor. Our results show that FSS stimulation rapidly induces phosphorylation of multiple TGFß family R-Smads by stimulating multimerization and concurrently activating several TGFß and BMP type I receptors, in a manner that requires the activity of the corresponding ligand. While the individual roles of the TGFß and BMP signaling pathways in bone mechanotransduction remain unclear, these results implicate that FSS activates both pathways to generate a downstream response that differs from that achieved by either ligand alone.


Subject(s)
Osteocytes/physiology , Receptor, Transforming Growth Factor-beta Type I/physiology , Activin Receptors, Type II/physiology , Animals , Cells, Cultured , Lab-On-A-Chip Devices , Mice , Protein Multimerization , Receptor, Transforming Growth Factor-beta Type I/chemistry , Sequence Analysis, RNA , Signal Transduction/physiology , Smad2 Protein/physiology , Smad3 Protein/physiology , Stress, Mechanical
2.
Dev Biol ; 442(1): 188-198, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30031756

ABSTRACT

Complex networks of signaling pathways maintain the correct balance between positive and negative growth signals, ensuring that tissues achieve proper sizes and differentiation pattern during development. In Drosophila, Dpp, a member of the TGFß family, plays two main roles during larval eye development. In the early eye primordium, Dpp promotes growth and cell survival, but later on, it switches its function to induce a developmentally-regulated cell cycle arrest in the G1 phase and neuronal photoreceptor differentiation. To advance in the identification and characterization of regulators and targets of Dpp signaling required for retinal development, we carried out an in vivo eye-targeted double-RNAi screen to identify punt (Type II TGFß receptor) interactors. Using a set of 251 genes associated with eye development, we identified CtBP, Dad, Ago and Brk as punt genetic interactors. Here, we show that downregulation of Ago, or conditions causing increased tissue growth including overexpression of Myc or CyclinD-Cdk4 are sufficient to partially rescue punt-dependent growth and photoreceptor differentiation. Interestingly, we show a novel role for the transcriptional co-repressor CtBP in inhibiting Dpp-dependent Mad activation by phosphorylation, downstream or in parallel to Dad, the inhibitory Smad. Furthermore, CtBP downregulation activates JNK signaling pathway, implying a complex regulation of signaling pathways by CtBP during eye development.


Subject(s)
Activin Receptors, Type II/physiology , Alcohol Oxidoreductases/physiology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Transcription Factors/metabolism , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Alcohol Oxidoreductases/metabolism , Animals , Cell Differentiation/genetics , Cyclin-Dependent Kinase 4 , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Eye/embryology , Eye/metabolism , Gene Expression Regulation, Developmental/genetics , Morphogenesis , Organogenesis , Repressor Proteins/metabolism , Signal Transduction/genetics , Transcription Factors/physiology , Transforming Growth Factor beta/metabolism
3.
Blood ; 120(20): 4263-73, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23018639

ABSTRACT

BMP9 signaling has been implicated in hereditary hemorrhagic telangiectasia (HHT) and vascular remodeling, acting via the HHT target genes, endoglin and ALK1. This study sought to identify endothelial BMP9-regulated proteins that could affect the HHT phenotype. Gene ontology analysis of cDNA microarray data obtained after BMP9 treatment of primary human endothelial cells indicated regulation of chemokine, adhesion, and inflammation pathways. These responses included the up-regulation of the chemokine CXCL12/SDF1 and down-regulation of its receptor CXCR4. Quantitative mass spectrometry identified additional secreted proteins, including the chemokine CXCL10/IP10. RNA knockdown of endoglin and ALK1 impaired SDF1/CXCR4 regulation by BMP9. Because of the association of SDF1 with ischemia, we analyzed its expression under hypoxia in response to BMP9 in vitro, and during the response to hindlimb ischemia, in endoglin-deficient mice. BMP9 and hypoxia were additive inducers of SDF1 expression. Moreover, the data suggest that endoglin deficiency impaired SDF1 expression in endothelial cells in vivo. Our data implicate BMP9 in regulation of the SDF1/CXCR4 chemokine axis in endothelial cells and point to a role for BMP9 signaling via endoglin in a switch from an SDF1-responsive autocrine phenotype to an SDF1 nonresponsive paracrine state that represses endothelial cell migration and may promote vessel maturation.


Subject(s)
Endothelial Cells/cytology , Growth Differentiation Factors/physiology , Neovascularization, Physiologic/physiology , Activin Receptors, Type I/physiology , Activin Receptors, Type II/physiology , Animals , Antigens, CD/physiology , Aorta/cytology , Autocrine Communication , Cell Hypoxia , Cell Movement , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/metabolism , Culture Media, Conditioned , Endoglin , Endothelial Cells/drug effects , Growth Differentiation Factor 2/pharmacology , Growth Differentiation Factor 2/physiology , Hindlimb/blood supply , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Ischemia/physiopathology , Mice , Paracrine Communication , RNA, Messenger/biosynthesis , RNA, Small Interfering/pharmacology , Real-Time Polymerase Chain Reaction , Receptors, Cell Surface/physiology , Transforming Growth Factor beta1/pharmacology
4.
Genesis ; 50(11): 783-800, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22611050

ABSTRACT

Hair follicles are simple, accessible models for many developmental processes. Here, using mutant mice, we show that Bmpr2, a known receptor for bone morphogenetic proteins (Bmps), and Acvr2a, a known receptor for Bmps and activins, are individually redundant but together essential for multiple follicular traits. When Bmpr2/Acvr2a function is reduced in cutaneous epithelium, hair follicles undergo rapid cycles of hair generation and loss. Alopecia results from a failure to terminate hair development properly, as hair clubs never form, and follicular retraction is slowed. Hair regeneration is rapid due to premature activation of new hair-production programs. Hair shafts differentiate aberrantly due to impaired arrest of medullary-cell proliferation. When Bmpr2/Acvr2a function is reduced in melanocytes, gray hair develops, as melanosomes differentiate but fail to grow, resulting in organelle miniaturization. We conclude that Bmpr2 and Acvr2a normally play cell-type-specific, necessary roles in organelle biogenesis and the shutdown of developmental programs and cell division.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/physiology , Hair Color , Hair/physiopathology , Activin Receptors, Type II/deficiency , Activin Receptors, Type II/genetics , Activin Receptors, Type II/physiology , Alopecia/genetics , Alopecia/metabolism , Animals , Bone Morphogenetic Protein Receptors, Type II/deficiency , Bone Morphogenetic Protein Receptors, Type II/genetics , Cell Differentiation , Cell Proliferation , Cells, Cultured , Female , Hair/growth & development , Hair/pathology , Hair Follicle/pathology , Male , Melanocytes/metabolism , Melanosomes/metabolism , Melanosomes/physiology , Mice , Mice, Transgenic , Primary Cell Culture
5.
Hum Reprod ; 26(12): 3346-54, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21984573

ABSTRACT

BACKGROUND: The Fallopian tube (FT) is the site of fertilization and early embryonic development. We have previously reported the expression of activins, their receptors and follistatin by the FT. Here, our aim was to study the expression of tubal activins, their type II receptors and follistatin during the menstrual cycle and following exposure to hCG in vivo. METHOD: A set of 30 FTs were collected from cycling women (n = 12) at different stages of the cycle (n = 4 in each stage) and pseudo-pregnant women (n = 3) at the time of hysterectomy for benign disease. The pseudo-pregnant women were injected with hCG in the days leading up to hysterectomy, and pseudo-pregnancy was confirmed by the persistence of amenorrhea, the presence of corpus luteum and decidualization of the endometrium. FT specimens were examined using immunohistochemistry and quantitative RT-PCR. RESULTS: The expression of activin ßA- and ßB-subunits, activin type IIA and IIB receptors, and follistatin varied throughout the menstrual cycle, being lowest in the follicular phase and highest in the luteal phase. These results were demonstrated at the mRNA and protein level by quantitative RT-PCR and immunohistochemistry (P< 0.05). HCG injection rescued the expression of the candidate molecules from falling to the follicular stage levels but the expression remained lower than in the luteal phase. CONCLUSIONS: We suggest that activins play a role in tubal physiology and early embryonic development. Additionally, exposure of the tubal epithelium to hCG modulates the expression of tubal activins.


Subject(s)
Activin Receptors, Type II/metabolism , Activins/metabolism , Fallopian Tubes/metabolism , Menstrual Cycle/metabolism , Pseudopregnancy/metabolism , Activin Receptors, Type II/physiology , Activins/physiology , Adult , Chorionic Gonadotropin/pharmacology , Female , Gene Expression Regulation/drug effects , Humans , Immunohistochemistry , Real-Time Polymerase Chain Reaction
6.
Muscle Nerve ; 43(5): 694-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21462203

ABSTRACT

INTRODUCTION: In this study we investigated the action of RAP-031, a soluble activin receptor type IIB (ActRIIB) comprised of a form of the ActRIIB extracellular domain linked to a murine Fc, and the NF-κB inhibitor, ursodeoxycholic acid (UDCA), on the whole body strength of mdx mice. METHODS: The whole body tension (WBT) method of assessing the forward pulling tension (FPT) exerted by dystrophic (mdx) mice was used. RESULTS: RAP-031 produced a 41% increase in body mass and a 42.5% increase in FPT without altering the FPT normalized for body mass (WBT). Coadministration of RAP-031 with UDCA produced increases in FPT that were associated with an increase in WBT. CONCLUSIONS: Myostatin inhibition increases muscle mass without altering the fundamental weakness characteristic of dystrophic muscle. Cotreatment with an NF-κB inhibitor potentiates the effects of myostatin inhibition in improving FPT in mdx mice.


Subject(s)
Activin Receptors, Type II/physiology , Muscle Tonus/physiology , Muscle, Skeletal/physiology , Muscular Dystrophy, Animal/physiopathology , Activin Receptors, Type II/pharmacology , Animals , Female , Male , Mice , Mice, Inbred mdx , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle Tonus/drug effects , Muscle, Skeletal/drug effects , Muscular Dystrophy, Animal/genetics , Solubility
7.
Mol Biol Cell ; 32(7): 605-621, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33566682

ABSTRACT

Complex formation and endocytosis of transforming growth factor-ß (TGF-ß) receptors play important roles in signaling. However, their interdependence remained unexplored. Here, we demonstrate that ALK1, a TGF-ß type I receptor prevalent in endothelial cells, forms stable complexes at the cell surface with endoglin and with type III TGF-ß receptors (TßRIII). We show that ALK1 undergoes clathrin-mediated endocytosis (CME) faster than ALK5, type II TGF-ß receptor (TßRII), endoglin, or TßRIII. These complexes regulate the endocytosis of the TGF-ß receptors, with a major effect mediated by ALK1. Thus, ALK1 enhances the endocytosis of TßRIII and endoglin, while ALK5 and TßRII mildly enhance endoglin, but not TßRIII, internalization. Conversely, the slowly endocytosed endoglin has no effect on the endocytosis of either ALK1, ALK5, or TßRII, while TßRIII has a differential effect, slowing the internalization of ALK5 and TßRII, but not ALK1. Such effects may be relevant to signaling, as BMP9-mediated Smad1/5/8 phosphorylation is inhibited by CME blockade in endothelial cells. We propose a model that links TGF-ß receptor oligomerization and endocytosis, based on which endocytosis signals are exposed/functional in specific receptor complexes. This has broad implications for signaling, implying that complex formation among various receptors regulates their surface levels and signaling intensities.


Subject(s)
Activin Receptors, Type II/metabolism , Endoglin/metabolism , Proteoglycans/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Activin Receptors, Type II/physiology , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Endocytosis , Endoglin/physiology , Endothelial Cells/metabolism , Humans , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Proteoglycans/physiology , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptor, Transforming Growth Factor-beta Type II/metabolism , Receptors, Transforming Growth Factor beta/physiology , Signal Transduction , Transforming Growth Factor beta/metabolism
8.
FASEB J ; 23(11): 3712-21, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19592636

ABSTRACT

ALK-1 is a transforming growth factor beta (TGF-beta) superfamily receptor that is predominantly expressed in endothelial cells and is essential for angiogenesis, as demonstrated by the embryonic lethal phentoype when targeted for deletion in mice and its mutation in the human disease hereditary hemorrhagic telangiectasia. Although ALK-1 and the endothelial-specific TGF-beta superfamily coreceptor, endoglin, form a heteromeric complex and bind similar TGF-beta superfamily ligands, their signaling mechanisms remain poorly characterized. Here we report the identification of CK2beta, the regulatory subunit of protein kinase CK2, as a novel enhancer of ALK-1 signaling. The cytoplasmic domain of ALK-1 specifically binds to CK2beta in vitro and in vivo. NAAIRS mutagenesis studies define amino acid sequences 181-199 of CK2beta and 207-212 of ALK-1 as the interaction domains, respectively. The ALK-1/CK2beta interaction specifically enhanced Smad1/5/8 phosphorylation and ALK-1-mediated reporter activation in response to TGF-beta1 and BMP-9 treatment. In a reciprocal manner, siRNA-mediated silencing of endogenous CK2beta inhibited TGF-beta1 and BMP-9-stimulated Smad1/5/8 phosphorylation and ALK-1-mediated reporter activation. Functionally, CK2beta enhanced the ability of activated or ligand-stimulated ALK-1 to inhibit endothelial cell migration. Similarly, ALK-1 and CK2beta antagonized endothelial tubule formation in Matrigel. These studies support CK2beta as an important regulator of ALK-1 signaling and ALK-1-mediated functions in endothelial cells.


Subject(s)
Activin Receptors, Type II/physiology , Casein Kinase II/metabolism , Signal Transduction/physiology , Animals , COS Cells , Chlorocebus aethiops , Humans , Protein Structure, Tertiary , Protein Subunits/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Two-Hybrid System Techniques
9.
Circ Res ; 102(8): 914-22, 2008 Apr 25.
Article in English | MEDLINE | ID: mdl-18309101

ABSTRACT

Angiogenesis is a complex process, requiring a finely tuned balance between numerous stimulatory and inhibitory signals. ALK1 (activin receptor like-kinase 1) is an endothelial-specific type 1 receptor of the transforming growth factor-beta receptor family. Heterozygotes with mutations in the ALK1 gene develop hereditary hemorrhagic telangiectasia type 2 (HHT2). Recently, we reported that bone morphogenetic protein (BMP)9 and BMP10 are specific ligands for ALK1 that potently inhibit microvascular endothelial cell migration and growth. These data lead us to suggest that these factors may play a role in the control of vascular quiescence. To test this hypothesis, we checked their presence in human serum. We found that human serum induced Smad1/5 phosphorylation. To identify the active factor, we tested neutralizing antibodies against BMP members and found that only the anti-BMP9 inhibited serum-induced Smad1/5 phosphorylation. The concentration of circulating BMP9 was found to vary between 2 and 12 ng/mL in sera and plasma from healthy humans, a value well above its EC(50) (50 pg/mL). These data indicated that BMP9 is circulating at a biologically active concentration. We then tested the effects of BMP9 in 2 in vivo angiogenic assays. We found that BMP9 strongly inhibited sprouting angiogenesis in the mouse sponge angiogenesis assay and that BMP9 could inhibit blood circulation in the chicken chorioallantoic membrane assay. Taken together, our results demonstrate that BMP9, circulating under a biologically active form, is a potent antiangiogenic factor that is likely to play a physiological role in the control of adult blood vessel quiescence.


Subject(s)
Activin Receptors, Type II/physiology , Bone Morphogenetic Proteins/physiology , Neovascularization, Physiologic , 3T3 Cells , Activin Receptors, Type II/genetics , Adult , Angiogenic Proteins , Animals , Bone Morphogenetic Proteins/blood , Case-Control Studies , Chick Embryo , Female , Growth Differentiation Factor 2 , Growth Differentiation Factors , Humans , Male , Mice , Middle Aged , Smad Proteins/metabolism , Telangiectasia, Hereditary Hemorrhagic/blood , Telangiectasia, Hereditary Hemorrhagic/genetics , Transfection
10.
Acta Biochim Biophys Sin (Shanghai) ; 42(10): 699-708, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20801928

ABSTRACT

Our previous studies have demonstrated that bone morphogenetic protein 9 (BMP-9) is one of the most efficacious BMPs to induce osteoblast differentiation of mesenchymal stem cells (MSCs). However, the molecular mechanism underlying the BMP-9-induced osteogenic differentiation of MSCs remains to be fully elucidated. In this study, dominant negative (DN) type II TGF-ß receptors were constructed and introduced into C3H10T1/2 stem cells, then in vitro and in vivo assays were carried out to analyze and identify the type II TGF-ß receptors required for BMP-9-induced osteogenesis. We found that three DN type II TGF-ß receptors, DN-BMPRII, DN-ActRII, and DN-ActRIIB, diminished BMP-9-induced alkaline phosphatase (ALP) activity, led to a decrease in BMP-9-induced Smad binding element (SBE)-controled reporter activity, reduced BMP-9-induced expressions of Smad6 and Smad7, and decreased BMP-9-induced mineralization in vitro and ectopic bone formation in vivo, finally resulted in decreased bone masses and immature osteogenesis. These findings strongly suggested that three wild-type II TGF-ß receptors, BMPRII, ActRII and ActRIIB, may play a functional role in BMP-9-induced osteogenic differentiation of C3H10T1/2 cells. However, C3H10T1/2 stem cells can express BMPRII and ActRII, but not ActRIIB. Using RNA interference (RNAi), we found that luciferase reporter activity and ALP activity induced by BMP-9 were accordingly inhibited along with the knockdown of BMPRII and ActRII. Taken together, our results demonstrated that BMPRII and ActRII are the functional type II TGF-ß receptors in BMP-9-induced osteogenic differentiation of C3H10T1/2 cells.


Subject(s)
Cell Differentiation/physiology , Growth Differentiation Factor 2/physiology , Mesenchymal Stem Cells/physiology , Protein Serine-Threonine Kinases/physiology , Receptors, Transforming Growth Factor beta/physiology , Activin Receptors, Type II/genetics , Activin Receptors, Type II/physiology , Alkaline Phosphatase/metabolism , Animals , Binding Sites/genetics , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/physiology , Cell Differentiation/genetics , Cell Line , Cell Transplantation , Growth Differentiation Factor 2/genetics , HCT116 Cells , HEK293 Cells , Humans , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Osteogenesis/genetics , Osteogenesis/physiology , Protein Serine-Threonine Kinases/genetics , RNA Interference , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Smad6 Protein/genetics , Smad6 Protein/metabolism , Smad7 Protein/genetics , Smad7 Protein/metabolism , Transfection
11.
Dev Dyn ; 238(9): 2374-81, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19653325

ABSTRACT

Bone morphogenetic protein-3 (BMP) has been identified as a negative regulator in the skeleton as mice lacking BMP3 have increased bone mass. To further understand how BMP3 mediates bone formation, we created transgenic mice overexpressing BMP3 using the type I collagen promoter. BMP3 transgenic mice displayed spontaneous rib fractures that were first detected at E17.0. The fractures were due to defects in differentiation of the periosteum and late hypertrophic chondrocytes resulting in thinner cortical bone with decreased mineralization. As BMP3 modulates BMP and activin signaling through ActRIIB, we examined the ribs of ActRIIB receptor knockout mice and found they had defects in late chondrogenesis and mineralization similar to BMP3 transgenic mice. These data suggest that BMP3 exerts its effects in the skeleton by altering signaling through ActRIIB in chondrocytes and the periosteum, and this results in defects in bone collar formation and late hypertrophic chondrocyte maturation leading to decreased mineralization and less bone.


Subject(s)
Bone Morphogenetic Protein 3/physiology , Fractures, Spontaneous/genetics , Rib Fractures/genetics , Activin Receptors, Type II/genetics , Activin Receptors, Type II/physiology , Animals , Blotting, Northern , Bone Morphogenetic Protein 3/genetics , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , Mice, Transgenic , Polymerase Chain Reaction
12.
Article in English | MEDLINE | ID: mdl-28348036

ABSTRACT

Genetic studies in animals and humans indicate that gene mutations that functionally perturb transforming growth factor ß (TGF-ß) signaling are linked to specific hereditary vascular syndromes, including Osler-Rendu-Weber disease or hereditary hemorrhagic telangiectasia and Marfan syndrome. Disturbed TGF-ß signaling can also cause nonhereditary disorders like atherosclerosis and cardiac fibrosis. Accordingly, cell culture studies using endothelial cells or smooth muscle cells (SMCs), cultured alone or together in two- or three-dimensional cell culture assays, on plastic or embedded in matrix, have shown that TGF-ß has a pivotal effect on endothelial and SMC proliferation, differentiation, migration, tube formation, and sprouting. Moreover, TGF-ß can stimulate endothelial-to-mesenchymal transition, a process shown to be of key importance in heart valve cushion formation and in various pathological vascular processes. Here, we discuss the roles of TGF-ß in vasculogenesis, angiogenesis, and lymphangiogenesis and the deregulation of TGF-ß signaling in cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/etiology , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Activin Receptors, Type II/physiology , Animals , Aortic Aneurysm/etiology , Atherosclerosis/etiology , Cardiovascular Diseases/physiopathology , Cell Communication , Endothelial Cells/physiology , Fibrosis , Humans , Hypertension, Pulmonary/etiology , Lymphangiogenesis , Myocardium/pathology , Neovascularization, Physiologic , Telangiectasia, Hereditary Hemorrhagic/etiology
13.
Sci Rep ; 7(1): 14470, 2017 10 31.
Article in English | MEDLINE | ID: mdl-29089584

ABSTRACT

Chemotherapy promotes the development of cachexia, a debilitating condition characterized by muscle and fat loss. ACVR2B/Fc, an inhibitor of the Activin Receptor 2B signaling, has been shown to preserve muscle mass and prolong survival in tumor hosts, and to increase bone mass in models of osteogenesis imperfecta and muscular dystrophy. We compared the effects of ACVR2B/Fc on muscle and bone mass in mice exposed to Folfiri. In addition to impairing muscle mass and function, Folfiri had severe negative effects on bone, as shown by reduced trabecular bone volume fraction (BV/TV), thickness (Tb.Th), number (Tb.N), connectivity density (Conn.Dn), and by increased separation (Tb.Sp) in trabecular bone of the femur and vertebra. ACVR2B/Fc prevented the loss of muscle mass and strength, and the loss of trabecular bone in femurs and vertebrae following Folfiri administration. Neither Folfiri nor ACVR2B/Fc had effects on femoral cortical bone, as shown by unchanged cortical bone volume fraction (Ct.BV/TV), thickness (Ct.Th) and porosity. Our results suggest that Folfiri is responsible for concomitant muscle and bone degeneration, and that ACVR2B/Fc prevents these derangements. Future studies are required to determine if the same protective effects are observed in combination with other anticancer regimens or in the presence of cancer.


Subject(s)
Activin Receptors, Type II/physiology , Bone Density/drug effects , Muscular Dystrophies/pathology , Activin Receptors, Type II/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bone and Bones , Cachexia/drug therapy , Camptothecin/adverse effects , Camptothecin/analogs & derivatives , Drug Therapy , Drug-Related Side Effects and Adverse Reactions/physiopathology , Female , Femur/drug effects , Fluorouracil/adverse effects , Induction Chemotherapy/methods , Leucovorin/adverse effects , Male , Mice , Mice, Inbred Strains , Muscle, Skeletal/pathology
14.
BMC Cell Biol ; 7: 16, 2006 Mar 29.
Article in English | MEDLINE | ID: mdl-16571110

ABSTRACT

BACKGROUND: In endothelial cells (EC), transforming growth factor-beta (TGF-beta) can bind to and transduce signals through ALK1 and ALK5. The TGF-beta/ALK5 and TGF-beta/ALK1 pathways have opposite effects on EC behaviour. Besides differential receptor binding, the duration of TGF-beta signaling is an important specificity determinant for signaling responses. TGF-beta/ALK1-induced Smad1/5 phosphorylation in ECs occurs transiently. RESULTS: The temporal activation of TGF-beta-induced Smad1/5 phosphorylation in ECs was found to be affected by de novo protein synthesis, and ALK1 and Smad5 expression levels determined signal strength of TGF-beta/ALK1 signaling pathway. Smad7 and protein phosphatase 1alpha (PP1alpha) mRNA expression levels were found to be specifically upregulated by TGF-beta/ALK1. Ectopic expression of Smad7 or PP1alpha potently inhibited TGF-beta/ALK1-induced Smad1/5 phosphorylation in ECs. Conversely, siRNA-mediated knockdown of Smad7 or PP1alpha enhanced TGF-beta/ALK1-induced signaling responses. PP1alpha interacted with ALK1 and this association was further potentiated by Smad7. Dephosphorylation of the ALK1, immunoprecipitated from cell lysates, was attenuated by a specific PP1 inhibitor. CONCLUSION: Our results suggest that upon its induction by the TGF-beta/ALK1 pathway, Smad7 may recruit PP1alpha to ALK1, and thereby control TGF-beta/ALK1-induced Smad1/5 phosphorylation.


Subject(s)
Activin Receptors, Type II/physiology , Endothelium, Vascular/physiology , Phosphoprotein Phosphatases/physiology , Signal Transduction/physiology , Smad7 Protein/physiology , Transforming Growth Factor beta/physiology , Activin Receptors/analysis , Activin Receptors/physiology , Activin Receptors, Type II/analysis , Adenoviridae/genetics , Animals , Blotting, Western , Cell Line , Endothelium, Vascular/chemistry , Endothelium, Vascular/cytology , Endothelium, Vascular/virology , Gene Expression Regulation/physiology , Immunoprecipitation , Mice , Phosphoprotein Phosphatases/analysis , Phosphoprotein Phosphatases/genetics , Phosphorylation , RNA, Messenger/analysis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Smad1 Protein/metabolism , Smad7 Protein/analysis , Smad7 Protein/genetics , Transcription, Genetic/physiology , Transfection , Transforming Growth Factor beta/analysis
15.
BMC Cardiovasc Disord ; 6: 13, 2006 Apr 04.
Article in English | MEDLINE | ID: mdl-16594992

ABSTRACT

BACKGROUND: TGF-beta1 is an important angiogenic factor involved in the different aspects of angiogenesis and vessel maintenance. TGF-beta signalling is mediated by the TbetaRII/ALK5 receptor complex activating the Smad2/Smad3 pathway. In endothelial cells TGF-beta utilizes a second type I receptor, ALK1, activating the Smad1/Smad5 pathway. Consequently, a perturbance of ALK1, ALK5 or TbetaRII activity leads to vascular defects. Mutations in ALK1 cause the vascular disorder hereditary hemorrhagic telangiectasia (HHT). METHODS: The identification of ALK1 and not ALK5 regulated genes in endothelial cells, might help to better understand the development of HHT. Therefore, the human microvascular endothelial cell line HMEC-1 was infected with a recombinant constitutively active ALK1 adenovirus, and gene expression was studied by using gene arrays and quantitative real-time PCR analysis. RESULTS: After 24 hours, 34 genes were identified to be up-regulated by ALK1 signalling. Analysing ALK1 regulated gene expression after 4 hours revealed 13 genes to be up- and 2 to be down-regulated. Several of these genes, including IL-8, ET-1, ID1, HPTPeta and TEAD4 are reported to be involved in angiogenesis. Evaluation of ALK1 regulated gene expression in different human endothelial cell types was not in complete agreement. Further on, disparity between constitutively active ALK1 and TGF-beta1 induced gene expression in HMEC-1 cells and primary HUVECs was observed. CONCLUSION: Gene array analysis identified 49 genes to be regulated by ALK1 signalling and at least 14 genes are reported to be involved in angiogenesis. There was substantial agreement between the gene array and quantitative real-time PCR data. The angiogenesis related genes might be potential HHT modifier genes. In addition, the results suggest endothelial cell type specific ALK1 and TGF-beta signalling.


Subject(s)
Activin Receptors, Type II/physiology , Gene Expression Regulation/genetics , Neovascularization, Physiologic/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , Transforming Growth Factor beta/physiology , Cell Line , Cells, Cultured , Constitutive Androstane Receptor , Endothelial Cells , Endothelium, Vascular/cytology , Humans , Transforming Growth Factor beta1
16.
Cancer Res ; 64(21): 7690-6, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15520171

ABSTRACT

The activin type II receptor (ACVR2) gene is a putative tumor suppressor gene that is frequently mutated in microsatellite-unstable colon cancers (MSI-H colon cancers). ACVR2 is a member of the transforming growth factor (TGF)-beta type II receptor (TGFBR2) family and controls cell growth and differentiation. SMAD proteins are major intracellular effectors shared by ACVR2 and TGFBR2 signaling; however, additional shared effector mechanisms remain to be explored. To discover novel mechanisms transmitting the ACVR2 signal, we restored ACVR2 function by transfecting wild-type ACVR2 (wt-ACVR2) into a MSI-H colon cancer cell line carrying an ACVR2 frameshift mutation. The effect of ACVR2 restoration on cell growth, SMAD phosphorylation, and global molecular phenotype was then evaluated. Decreased cell growth was observed in wt-ACVR2 transfectants relative to ACVR2-deficient vector-transfected controls. Western blotting revealed higher expression of phosphorylated SMAD2 in wt-ACVR2 transfectants versus controls, suggesting cells deficient in ACVR2 had impaired SMAD signaling. Microarray-based differential expression analysis revealed substantial ACVR2-induced overexpression of genes implicated in the control of cell growth and tumorigenesis, including the activator protein (AP)-1 complex genes JUND, JUN, and FOSB, as well as the small GTPase signal transduction family members, RHOB, ARHE, and ARHGDIA. Overexpression of these genes is shared with TGFBR2 activation. This observed similarity between the activin and TGF-beta signaling systems suggests that activin may serve as an alternative activator of TGF-beta effectors, including SMADs, and that frameshift mutation of ACVR2 may contribute to MSI-H colon tumorigenesis via disruption of alternate TGF-beta effector pathways.


Subject(s)
Activin Receptors, Type II/physiology , Colonic Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Signal Transduction , Transforming Growth Factor beta/pharmacology , Cell Division , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Humans , Phosphorylation , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/physiology , Smad2 Protein , Trans-Activators/metabolism
17.
Stem Cells Dev ; 24(3): 312-9, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25203845

ABSTRACT

microRNAs (miRNAs) are short 20- to 22-nucleotide noncoding RNAs that negatively regulate the expression of target genes at the post-transcriptional level. The expression of specific miRNAs and their roles in the osteogenic differentiation of human periodontal ligament stem cells (PDLSCs) exposed to mechanical stretch remain unclear. Here, we found that stretch induced both osteogenic differentiation and the differential expression of miR-21 in PDLSCs. Furthermore, we identified activin receptor type IIB (ACVR2B) as a target gene of miR-21. Luciferase reporter assays showed that miR-21 interacts directly with the 3'-untranslated repeat sequence of ACVR2B mRNA. Mechanical stretch suppressed ACVR2B protein levels in PDLSCs, and this suppressive effect was modulated when endogenous miR-21 levels were either enhanced or inhibited. Both stretch and the expression of miR-21 altered endogenous ACVR2B protein levels and thus the osteogenic differentiation of PDLSCs. In addition, gain- and loss of function of ACVR2B mediated the osteogenic differentiation of PDLSCs. This study demonstrates that miR-21 is a mechanosensitive gene that plays an important role in the osteogenic differentiation of PDLSCs exposed to stretch.


Subject(s)
Activin Receptors, Type II/physiology , Gene Expression Regulation, Developmental , MicroRNAs/physiology , Multipotent Stem Cells/cytology , Osteogenesis/physiology , Periodontal Ligament/cytology , Stress, Mechanical , 3' Untranslated Regions , Activin Receptors, Type II/biosynthesis , Activin Receptors, Type II/genetics , Adolescent , Bone Remodeling/physiology , Cells, Cultured , Child , Gene Regulatory Networks , Genetic Vectors , Humans , Multipotent Stem Cells/metabolism , Osteogenesis/genetics , RNA/genetics , RNA/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Transduction, Genetic , Transforming Growth Factor beta/physiology
18.
Mol Cell Endocrinol ; 211(1-2): 105-13, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-14656483

ABSTRACT

Bone morphogenetic proteins (BMPs) are multifunctional proteins that regulate the fate of different cell types, including mesenchymal and endothelial cells. BMPs inhibit myogenic differentiation, but promote the differentiation of mesenchymal cells into osteoblasts. Furthermore, endothelial migration and tube formation are stimulated by BMPs. Like other members of the transforming growth factor-beta (TGF-beta) superfamily, BMPs elicit their cellular effects via specific types I and II serine/threonine receptors. The activated BMP type I receptor phosphorylates specific receptor-regulated (R)-Smad proteins, which assemble into heteromeric complexes with common partner (Co)-Smad4. Heteromeric Smad complexes efficiently translocate into the nucleus, where they regulate the transcription of target genes. Inhibitors of differentiation (Id) are genes that are specifically induced by BMPs in tissues of different origin. Promoter analysis of Id1 indicates three distinct sequence elements that are sufficient and essential for efficient BMP-induced activation. Furthermore, recent studies reveal an important effector function for Id1 in various BMP-induced biological responses.


Subject(s)
Cell Differentiation/physiology , Receptors, Growth Factor/physiology , Repressor Proteins , Activin Receptors, Type I/physiology , Activin Receptors, Type II/physiology , Animals , Bone Morphogenetic Protein Receptors , Bone Morphogenetic Protein Receptors, Type I , Bone Morphogenetic Protein Receptors, Type II , Bone Morphogenetic Proteins/physiology , DNA-Binding Proteins/physiology , Humans , Inhibitor of Differentiation Protein 1 , Models, Biological , Phosphoproteins/physiology , Protein Serine-Threonine Kinases/physiology , Signal Transduction/physiology , Smad Proteins , Smad4 Protein , Smad5 Protein , Trans-Activators/physiology , Transcription Factors/physiology
19.
APMIS ; 111(1): 174-81; discussion 182-3, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12760377

ABSTRACT

In humans, Sertoli cell tumors account for approximately 4% of all testicular tumors, and 20% of these are malignant. The mechanisms underlying Sertoli cell tumorigenesis remain largely unknown. Using gene knockout technology, we previously generated mutant mice lacking the alpha subunit of inhibin dimers. The inhibin alpha-null male mice develop testicular Sertoli cell tumors with 100% penetrance. These tumors develop as early as 4 weeks of age and cause a cachexia-like wasting syndrome. Castrated inhibin alpha knockout mice develop sex steroidogenic adrenal cortical tumors. These studies have identified inhibins as secreted tumor suppressors with specificity for the gonads and adrenal glands. It had been suggested that endocrine factors play roles in Sertoli cell tumorigenesis by altering cell cycle machinery of the Sertoli cells. To test the potential of these factors to function as modifiers of Sertoli cell tumorigenesis, we have employed a genetic intercross strategy, breeding inhibin a mutant mice with mutant mice deficient in endocrine signaling factors including gonadotropin releasing hormone (hypogonadal, hpg mice), follicle stimulating hormone, anti-Miillerian hormone (AMH), activin receptor type II, or androgen receptor (testicular feminization, tfm mice), or mice overexpressing follistatin. We are also investigating the effects of loss of critical cell cycle regulators, such as cyclin dependent kinase inhibitor p27, on Sertoli cell tumorigenesis in inhibin alpha knockout males. These studies clearly demonstrate the roles of these factors as modifiers of the Sertoli cell tumorigenesis. Activin signaling through activin receptor type II is responsible for the cachexia-like syndrome observed in the inhibin a knockout mice with tumors. The gonadotropin hormones are essential for testicular tumor development, but elevated FSH levels are not sufficient to cause Sertoli cell tumors. Absence of FSH, lack of androgen receptor, or overexpression of follistatin slows the tumor growth and minimizes the cachexia symptoms, thus prolonging the life span of these double mutant mice. In contrast, absence of AMH or p27 causes earlier onset and more aggressive development of testicular tumor, with an earlier death of double mutant mice. We are currently investigating roles of estrogen signaling pathways, and other cell cycle regulators, in tumor development in the inhibin alpha knockout mice by generating mice with double or triple mutations. Genetic engineering in mouse models provides a powerful tool to study the mechanisms of testicular tumorigenesis and define the important genetic modifiers in vivo.


Subject(s)
Sertoli Cell Tumor/etiology , Testicular Neoplasms/etiology , Activin Receptors, Type II/physiology , Animals , Cachexia/physiopathology , Cell Cycle , Gene Deletion , Genetic Engineering , Gonadotropins, Pituitary/metabolism , Humans , Inhibins/genetics , Inhibins/physiology , Male , Mice , Mice, Knockout , Models, Animal , Sertoli Cell Tumor/physiopathology , Testicular Neoplasms/physiopathology , Testis/metabolism , Testis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL