Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Cell Signal ; 118: 111142, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38508350

ABSTRACT

OBJECTIVE: To elucidate the molecular mechanism of overloading-induced osteoarthritis (OA) and to find a novel therapeutic target. METHODS: We utilized human cartilage specimens, mouse chondrocytes, a destabilization of the medial meniscus (DMM) mouse model, and a mouse hindlimb weight-bearing model to validate the role of overloading on chondrocyte senescence and OA development. Then, we observed the effect of PIEZO1-miR-155-5p-GDF6-SMAD2/3 signaling axis on the preservation of joint metabolic homeostasis under overloading in vivo, in vitro and ex vivo by qPCR, Western blot, enzyme-linked immunosorbent assay (ELISA), immunohistochemistry, immunofluorescence, SA-ß-gal staining, CCK8 assay, et al. Finally, we verified the therapeutic effects of intra-articular injection of miR-155-5p inhibitor or recombinant GDF6 on the murine overloading-induced OA models. RESULTS: Chondrocytes sensesed the mechanical overloading through PIEZO1 and up-regulated miR-155-5p expression. MiR-155-5p mimics could copy the effects of overloading-induced chondrocyte senescence and OA. Additionally, miR-155-5p could suppress the mRNA expression of Gdf6-Smad2/3 in various tissues within the joint. Overloading could disrupt joint metabolic homeostasis by downregulating the expression of anabolism indicators and upregulating the expression of catabolism indicators in the chondrocytes and synoviocytes, while miR-155-5p inhibition or GDF6 supplementation could exert an antagonistic effect by preserving the joint homeostasis. Finally, in the in vivo overloading models, intra-articular injection of miR-155-5p inhibitor or recombinant GDF6 could significantly mitigate the severity of impending OA and lessened the progression of existing OA. CONCLUSION: GDF6 overexpression or miR-155-5p inhibition could attenuate overloading-induced chondrocyte senescence and OA through the PIEZO1-miR-155-5p-GDF6-SMAD2/3 signaling pathway. Our study provides a new therapeutic target for the treatment of overloading-induced OA.


Subject(s)
MicroRNAs , Osteoarthritis , Animals , Humans , Mice , Apoptosis , Chondrocytes/metabolism , Growth Differentiation Factor 6/metabolism , Growth Differentiation Factor 6/pharmacology , Growth Differentiation Factor 6/therapeutic use , Ion Channels/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Osteoarthritis/metabolism , Signal Transduction , Smad2 Protein/metabolism , Stress, Mechanical
2.
J Bone Miner Res ; 38(4): 568-577, 2023 04.
Article in English | MEDLINE | ID: mdl-36744814

ABSTRACT

Multiple synostoses syndromes (SYNS) are a group of rare genetic bone disorders characterized by multiple joint fusions. We previously reported an SYNS4-causing GDF6 c.1330 T > A (p.Tyr444Asn) mutation, which reduced Noggin-induced GDF6 inhibition and enhanced SMAD1/5/8 signaling. However, the mechanisms by which GDF6 gain-of-function mutation alters joint formation and the comprehensive molecular portraits of SYNS4 remain unclear. Herein, we introduce the p.Tyr443Asn (orthologous to the human GDF6 p.Tyr444Asn) mutation into the mouse Gdf6 locus and report the results of extensive phenotype analysis, joint development investigation, and transcriptome profiling of Gdf6 p.Tyr443Asn limb buds. Gdf6 p.Tyr443Asn knock-in mice recapitulated the morphological features of human SYNS4, showing joint fusion in the wrists, ankles, phalanges, and auditory ossicles. Analysis of mouse embryonic forelimbs demonstrated joint interzone formation defects and excess chondrogenesis in Gdf6 p.Tyr443Asn knock-in mice. Further, RNA sequencing of forelimb buds revealed enhanced bone formation and upregulated bone morphogenetic protein (BMP) signaling in mice carrying the Gdf6 p.Tyr443Asn mutation. Because tightly regulated BMP signaling is critical for skeletal development and joint morphogenesis, our study shows that enhancing GDF6 activity has a significant impact on both prenatal joint development and postnatal joint maintenance. © 2023 American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Bone Morphogenetic Proteins , Growth Differentiation Factor 6 , Synostosis , Animals , Humans , Mice , Bone and Bones/metabolism , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Growth Differentiation Factor 6/genetics , Growth Differentiation Factor 6/metabolism , Mutation/genetics , Synostosis/genetics
3.
Genes (Basel) ; 13(7)2022 07 04.
Article in English | MEDLINE | ID: mdl-35885978

ABSTRACT

The human capacity to speak is fundamental to our advanced intellectual, technological and social development. Yet so very little is known regarding the evolutionary genetics of speech or its relationship with the broader aspects of evolutionary development in primates. In this study, we describe a large family with evolutionary retrograde development of the larynx and wrist. The family presented with severe speech impairment and incremental retrograde elongations of the pisiform in the wrist that limited wrist rotation from 180° to 90° as in primitive primates. To our surprise, we found that a previously unknown primate-specific gene TOSPEAK had been disrupted in the family. TOSPEAK emerged de novo in an ancestor of extant primates across a 540 kb region of the genome with a pre-existing highly conserved long-range laryngeal enhancer for a neighbouring bone morphogenetic protein gene GDF6. We used transgenic mouse modelling to identify two additional GDF6 long-range enhancers within TOSPEAK that regulate GDF6 expression in the wrist. Disruption of TOSPEAK in the affected family blocked the transcription of TOSPEAK across the 3 GDF6 enhancers in association with a reduction in GDF6 expression and retrograde development of the larynx and wrist. Furthermore, we describe how TOSPEAK developed a human-specific promoter through the expansion of a penta-nucleotide direct repeat that first emerged de novo in the promoter of TOSPEAK in gibbon. This repeat subsequently expanded incrementally in higher hominids to form an overlapping series of Sp1/KLF transcription factor consensus binding sites in human that correlated with incremental increases in the promoter strength of TOSPEAK with human having the strongest promoter. Our research indicates a dual evolutionary role for the incremental increases in TOSPEAK transcriptional interference of GDF6 enhancers in the incremental evolutionary development of the wrist and larynx in hominids and the human capacity to speak and their retrogression with the reduction of TOSPEAK transcription in the affected family.


Subject(s)
Growth Differentiation Factor 6 , Speech , Animals , Biological Evolution , Growth Differentiation Factor 6/genetics , Growth Differentiation Factor 6/metabolism , Humans , Mice , Primates/genetics , Regulatory Sequences, Nucleic Acid
4.
Cells ; 11(7)2022 03 31.
Article in English | MEDLINE | ID: mdl-35406739

ABSTRACT

Growth differentiation factors (GDFs) regulate homeostasis by amplifying extracellular matrix anabolism and inhibiting pro-inflammatory cytokine production in the intervertebral disc (IVD). The aim of this study was to elucidate the effects of GDF-6 on human IVD nucleus pulposus (NP) cells using a three-dimensional culturing system in vitro and on rat tail IVD tissues using a puncture model in vivo. In vitro, Western blotting showed decreased GDF-6 expression with age and degeneration severity in surgically collected human IVD tissues (n = 12). Then, in moderately degenerated human IVD NP cells treated with GDF-6 (100 ng/mL), immunofluorescence demonstrated an increased expression of matrix components including aggrecan and type II collagen. Quantitative polymerase chain reaction analysis also presented GDF-6-induced downregulation of pro-inflammatory tumor necrosis factor (TNF)-α (p = 0.014) and interleukin (IL)-6 (p = 0.016) gene expression stimulated by IL-1ß (10 ng/mL). Furthermore, in the mitogen-activated protein kinase pathway, Western blotting displayed GDF-6-induced suppression of p38 phosphorylation (p = 0.041) under IL-1ß stimulation. In vivo, intradiscal co-administration of GDF-6 and atelocollagen was effective in alleviating rat tail IVD annular puncture-induced radiologic height loss (p = 0.005), histomorphological degeneration (p < 0.001), matrix metabolism (aggrecan, p < 0.001; type II collagen, p = 0.001), and pro-inflammatory cytokine production (TNF-α, p < 0.001; IL-6, p < 0.001). Consequently, GDF-6 could be a therapeutic growth factor for degenerative IVD disease.


Subject(s)
Growth Differentiation Factor 6 , Intervertebral Disc Degeneration , Intervertebral Disc , Aggrecans/metabolism , Animals , Collagen Type II/metabolism , Growth Differentiation Factor 6/metabolism , Humans , Intervertebral Disc/pathology , Intervertebral Disc Degeneration/metabolism , Rats , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
5.
Adv Sci (Weinh) ; 8(18): e2004629, 2021 09.
Article in English | MEDLINE | ID: mdl-34319658

ABSTRACT

Angiogenesis is essential for vascular development. The roles of regulatory long noncoding RNAs (lncRNAs) in mediating angiogenesis remain under-explored. Human embryonic stem cell-derived mesenchymal stem cells (hES-MSCs) are shown to exert more potent cardioprotective effects against cardiac ischemia than human bone marrow-derived MSCs (hBM-MSCs), associated with enhanced neovascularization. The purpose of this study is to search for angiogenic lncRNAs enriched in hES-MSCs, and investigate their roles and mechanisms. AC103746.1 is one of the most highly expressed intergenic lncRNAs detected in hES-MSCs versus hBM-MSCs, and named as SCDAL (stem cell-derived angiogenic lncRNA). SCDAL knockdown significantly reduce the angiogenic potential and reparative effects of hES-MSCs in the infarcted hearts, while overexpression of SCDAL in either hES-MSCs or hBM-MSCs exhibits augmented angiogenesis and cardiac function recovery. Mechanistically, SCDAL induces growth differentiation factor 6 (GDF6) expression via direct interaction with SNF5 at GDF6 promoter. Secreted GDF6 promotes endothelial angiogenesis via non-canonical vascular endothelial growth factor receptor 2 activation. Furthermore, SCDAL-GDF6 is expressed in human endothelial cells, and directly enhances endothelial angiogenesis in vitro and in vivo. Thus, these findings uncover a previously unknown lncRNA-dependent regulatory circuit for angiogenesis. Targeted intervention of the SCDAL-GDF6 pathway has potential as a therapy for ischemic heart diseases.


Subject(s)
Growth Differentiation Factor 6/genetics , Growth Differentiation Factor 6/metabolism , Neovascularization, Pathologic/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , SMARCB1 Protein/genetics , SMARCB1 Protein/metabolism , Adult , Female , Gene Expression/genetics , Humans , Male , Middle Aged , Neovascularization, Pathologic/metabolism , Signal Transduction/genetics
6.
Arterioscler Thromb Vasc Biol ; 41(2): 698-710, 2021 02.
Article in English | MEDLINE | ID: mdl-33054395

ABSTRACT

OBJECTIVE: The superoxide-generating Nox2 (NADPH oxidase-2) is expressed in multiple cell types. Previous studies demonstrated distinct roles for cardiomyocyte, endothelial cell, and leukocyte cell Nox2 in ANG II (angiotensin II)-induced cardiovascular remodeling. However, the in vivo role of fibroblast Nox2 remains unclear. Approach and Results: We developed a novel mouse model with inducible fibroblast-specific deficiency of Nox2 (fibroblast-specific Nox2 knockout or Fibro-Nox2KO mice) and investigated the responses to chronic ANG II stimulation. Fibro-Nox2KO mice showed no differences in basal blood pressure or vessel wall morphology, but the hypertensive response to ANG II infusion (1.1 mg/[kg·day] for 14 days) was substantially reduced as compared to control Nox2-Flox littermates. This was accompanied by a significant attenuation of aortic and resistance vessel remodeling. The conditioned medium of ANG II-stimulated primary fibroblasts induced a significant increase in vascular smooth muscle cell growth, which was inhibited by the short hairpin RNA (shRNA)-mediated knockdown of fibroblast Nox2. Mass spectrometric analysis of the secretome of ANG II-treated primary fibroblasts identified GDF6 (growth differentiation factor 6) as a potential growth factor that may be involved in these effects. Recombinant GDF6 induced a concentration-dependent increase in vascular smooth muscle cell growth while chronic ANG II infusion in vivo significantly increased aortic GDF6 protein levels in control mice but not Fibro-Nox2KO animals. Finally, silencing GDF6 in fibroblasts prevented the induction of vascular smooth muscle cell growth by fibroblast-conditioned media in vitro. CONCLUSIONS: These results indicate that fibroblast Nox2 plays a crucial role in the development of ANG II-induced vascular remodeling and hypertension in vivo. Mechanistically, fibroblast Nox2 may regulate paracrine signaling to medial vascular smooth muscle cells via factors, such as GDF6.


Subject(s)
Fibroblasts/enzymology , Hypertension/enzymology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , NADPH Oxidase 2/metabolism , Paracrine Communication , Vascular Remodeling , Angiotensin II , Animals , Aorta/metabolism , Aorta/pathology , Aorta/physiopathology , Blood Pressure , Cells, Cultured , Disease Models, Animal , Growth Differentiation Factor 6/genetics , Growth Differentiation Factor 6/metabolism , Hypertension/chemically induced , Hypertension/genetics , Hypertension/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/pathology , NADPH Oxidase 2/genetics , Signal Transduction
7.
Cell Rep ; 33(5): 108332, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33147457

ABSTRACT

We report here that the autocrine signaling mediated by growth and differentiation factor 6 (GDF6), a member of the bone morphogenetic protein (BMP) family of cytokines, maintains Ewing sarcoma growth by preventing Src hyperactivation. Surprisingly, Ewing sarcoma depends on the prodomain, not the BMP domain, of GDF6. We demonstrate that the GDF6 prodomain is a ligand for CD99, a transmembrane protein that has been widely used as a marker of Ewing sarcoma. The binding of the GDF6 prodomain to the CD99 extracellular domain results in recruitment of CSK (C-terminal Src kinase) to the YQKKK motif in the intracellular domain of CD99, inhibiting Src activity. GDF6 silencing causes hyperactivation of Src and p21-dependent growth arrest. We demonstrate that two GDF6 prodomain mutants linked to Klippel-Feil syndrome are hyperactive in CD99-Src signaling. These results reveal a cytokine signaling pathway that regulates the CSK-Src axis and cancer cell proliferation and suggest the gain-of-function activity for disease-causing GDF6 mutants.


Subject(s)
12E7 Antigen/metabolism , Growth Differentiation Factor 6/metabolism , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Signal Transduction , src-Family Kinases/metabolism , Animals , CSK Tyrosine-Protein Kinase/metabolism , Cell Proliferation , Down-Regulation , Gene Expression Regulation, Neoplastic , Growth Differentiation Factor 6/chemistry , Humans , Klippel-Feil Syndrome/genetics , Mice, SCID , Mutation/genetics , Oncogene Proteins, Fusion/metabolism , Protein Domains , Proteome/metabolism , Proteomics , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/metabolism , Transcription, Genetic
8.
Eur J Hum Genet ; 28(12): 1681-1693, 2020 12.
Article in English | MEDLINE | ID: mdl-32737436

ABSTRACT

Although over 50 genes are known to cause renal malformation if mutated, the underlying genetic basis, most easily identified in syndromic cases, remains unsolved in most patients. In search of novel causative genes, whole-exome sequencing in a patient with renal, i.e., crossed fused renal ectopia, and extrarenal, i.e., skeletal, eye, and ear, malformations yielded a rare heterozygous variant in the GDF6 gene encoding growth differentiation factor 6, a member of the BMP family of ligands. Previously, GDF6 variants were reported to cause pleiotropic defects including skeletal, e.g., vertebral, carpal, tarsal fusions, and ocular, e.g., microphthalmia and coloboma, phenotypes. To assess the role of GDF6 in the pathogenesis of renal malformation, we performed targeted sequencing in 193 further patients identifying rare GDF6 variants in two cases with kidney hypodysplasia and extrarenal manifestations. During development, gdf6 was expressed in the pronephric tubule of Xenopus laevis, and Gdf6 expression was observed in the ureteric tree of the murine kidney by RNA in situ hybridization. CRISPR/Cas9-derived knockout of Gdf6 attenuated migration of murine IMCD3 cells, an effect rescued by expression of wild-type but not mutant GDF6, indicating affected variant function regarding a fundamental developmental process. Knockdown of gdf6 in Xenopus laevis resulted in impaired pronephros development. Altogether, we identified rare heterozygous GDF6 variants in 1.6% of all renal anomaly patients and 5.4% of renal anomaly patients additionally manifesting skeletal, ocular, or auricular abnormalities, adding renal hypodysplasia and fusion to the phenotype spectrum of GDF6 variant carriers and suggesting an involvement of GDF6 in nephrogenesis.


Subject(s)
Growth Differentiation Factor 6/genetics , Urogenital Abnormalities/genetics , Vesico-Ureteral Reflux/genetics , Adolescent , Adult , Animals , Cell Line , Child , Child, Preschool , Female , Growth Differentiation Factor 6/metabolism , Heterozygote , Humans , Infant , Kidney Tubules/abnormalities , Kidney Tubules/metabolism , Male , Mice , Mutation , Urogenital Abnormalities/pathology , Vesico-Ureteral Reflux/pathology , Xenopus
9.
Reproduction ; 159(5): 525-537, 2020 05.
Article in English | MEDLINE | ID: mdl-32045359

ABSTRACT

Missed abortion (MA) is a common disease in obstetrics and gynecology. More and more studies have focused on the relationship between miRNAs and pregnancy maintenance and its related diseases. The aim of this article is to explore the relationship between miRNA and MA. The expression of miR-98 were detected by in situ hybridization and real-time PCR. Cell proliferation, activity and migration were measured via Edu, MTT, and transwell assays. The target genes of miR-98 are identified by dual-luciferase activity assay. And the expression levels of target genes were determined by Western blot, real-time PCR and immunohistochemistry. miR-98 was significantly up-regulated in placental villi from over 35 years old MA patients compared with the age-matched normal pregnant women. Up-regulation of miR-98 suppressed the proliferation, activity and migration of the human trophoblast HTR-8/SVneo cell in vitro. miR-98 could bind to GDF6 and FAPP2 mRNA 3'-UTR and negatively regulate their expression. The downregulation of miR-98 promoted cell proliferation, then knockdown of GDF6 or FAPP2 inhibited miR-98-mediated cell proliferation. GDF6 and FAPP2 expression in the placental villi from MA patients were decreased compared to normal placental tissues. The expression of miR-98 in MA had an opposite relationship with the expression of GDF6 and FAPP2. Overexpression of miR-98 is associated with the occurrence of MA. miR-98 prevents proliferation, viability and migration of trophoblast cells partially through targeting GDF6 and FAPP2.


Subject(s)
Abortion, Missed/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Movement/physiology , Cell Proliferation/physiology , Growth Differentiation Factor 6/metabolism , MicroRNAs/metabolism , Trophoblasts/metabolism , Abortion, Missed/genetics , Adaptor Proteins, Signal Transducing/genetics , Adult , Cell Line , Female , Growth Differentiation Factor 6/genetics , Humans , MicroRNAs/genetics , Placenta/metabolism , Pregnancy , Up-Regulation
10.
Elife ; 82019 02 18.
Article in English | MEDLINE | ID: mdl-30775968

ABSTRACT

We use the transparency of zebrafish embryos to reveal the de novo generation of a simple squamous epithelium and identify the cellular architecture in the epithelial transition zone that ties this squamous epithelium to the columnar neuroepithelium within the embryo's brain. The simple squamous epithelium of the rhombencephalic roof plate is pioneered by distinct mesenchymal cells at the dorsal midline of the neural tube. Subsequently, a progenitor zone is established at the interface between columnar epithelium of the rhombic lip and the expanding squamous epithelium of the roof plate. Surprisingly, this interface consists of a single progenitor cell type that we have named the veil cell. Veil cells express gdf6a and constitute a lineage restricted stem zone that generates the squamous roof plate by direct transformation and asymmetrically fated divisions. Experimental restriction of roof plate expansion leads to extrusion of veil cell daughters and squamous cells, suggesting veil cell fate is regulated by the space available for roof plate growth.


Subject(s)
Cerebral Ventricles/anatomy & histology , Epithelium/anatomy & histology , Zebrafish/anatomy & histology , Animals , Asymmetric Cell Division , Cell Proliferation , Cell Self Renewal , Cerebral Ventricles/cytology , Embryo, Nonmammalian/cytology , Epithelium/embryology , Growth Differentiation Factor 6/metabolism , Mesoderm/embryology , Rhombencephalon/anatomy & histology , Rhombencephalon/embryology , Zebrafish/embryology , Zebrafish Proteins/metabolism
11.
PLoS Genet ; 14(3): e1007246, 2018 03.
Article in English | MEDLINE | ID: mdl-29522511

ABSTRACT

The eye primordium arises as a lateral outgrowth of the forebrain, with a transient fissure on the inferior side of the optic cup providing an entry point for developing blood vessels. Incomplete closure of the inferior ocular fissure results in coloboma, a disease characterized by gaps in the inferior eye and recognized as a significant cause of pediatric blindness. Here, we identify eight patients with defects in tissues of the superior eye, a congenital disorder that we term superior coloboma. The embryonic origin of superior coloboma could not be explained by conventional models of eye development, leading us to reanalyze morphogenesis of the dorsal eye. Our studies revealed the presence of the superior ocular sulcus (SOS), a transient division of the dorsal eye conserved across fish, chick, and mouse. Exome sequencing of superior coloboma patients identified rare variants in a Bone Morphogenetic Protein (Bmp) receptor (BMPR1A) and T-box transcription factor (TBX2). Consistent with this, we find sulcus closure defects in zebrafish lacking Bmp signaling or Tbx2b. In addition, loss of dorsal ocular Bmp is rescued by concomitant suppression of the ventral-specific Hedgehog pathway, arguing that sulcus closure is dependent on dorsal-ventral eye patterning cues. The superior ocular sulcus acts as a conduit for blood vessels, with altered sulcus closure resulting in inappropriate connections between the hyaloid and superficial vascular systems. Together, our findings explain the existence of superior coloboma, a congenital ocular anomaly resulting from aberrant morphogenesis of a developmental structure.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I/genetics , Coloboma/embryology , Coloboma/genetics , Cytochrome P-450 CYP1B1/genetics , Eye/embryology , Adult , Animals , Animals, Genetically Modified , Bone Morphogenetic Protein Receptors, Type I/metabolism , Chick Embryo , Embryo, Nonmammalian , Growth Differentiation Factor 6/genetics , Growth Differentiation Factor 6/metabolism , Humans , Infant , Mice , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
12.
J Clin Invest ; 128(1): 294-308, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29202482

ABSTRACT

Oncogenomic studies indicate that copy number variation (CNV) alters genes involved in tumor progression; however, identification of specific driver genes affected by CNV has been difficult, as these rearrangements are often contained in large chromosomal intervals among several bystander genes. Here, we addressed this problem and identified a CNV-targeted oncogene by performing comparative oncogenomics of human and zebrafish melanomas. We determined that the gene encoding growth differentiation factor 6 (GDF6), which is the ligand for the BMP family, is recurrently amplified and transcriptionally upregulated in melanoma. GDF6-induced BMP signaling maintained a trunk neural crest gene signature in melanomas. Additionally, GDF6 repressed the melanocyte differentiation gene MITF and the proapoptotic factor SOX9, thereby preventing differentiation, inhibiting cell death, and promoting tumor growth. GDF6 was specifically expressed in melanomas but not melanocytes. Moreover, GDF6 expression levels in melanomas were inversely correlated with patient survival. Our study has identified a fundamental role for GDF6 and BMP signaling in governing an embryonic cell gene signature to promote melanoma progression, thus providing potential opportunities for targeted therapy to treat GDF6-positive cancers.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Growth Differentiation Factor 6/metabolism , Melanoma/metabolism , Neoplasm Proteins/metabolism , Signal Transduction , Animals , Bone Morphogenetic Proteins/genetics , Cell Line, Tumor , Female , Growth Differentiation Factor 6/genetics , HEK293 Cells , Humans , Ligands , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Neoplasm Proteins/genetics
13.
Arterioscler Thromb Vasc Biol ; 38(2): 353-362, 2018 02.
Article in English | MEDLINE | ID: mdl-29284606

ABSTRACT

OBJECTIVE: The assembly of a functional vascular system requires a coordinated and dynamic transition from activation to maturation. High vascular endothelial growth factor activity promotes activation, including junction destabilization and cell motility. Maturation involves junctional stabilization and formation of a functional endothelial barrier. The identity and mechanism of action of prostabilization signals are still mostly unknown. Bone morphogenetic protein receptors and their ligands have important functions during embryonic vessel assembly and maturation. Previous work has suggested a role for growth differentiation factor 6 (GDF6; bone morphogenetic protein 13) in vascular integrity although GDF6's mechanism of action was not clear. Therefore, we sought to further explore the requirement for GDF6 in vascular stabilization. APPROACH AND RESULTS: We investigated the role of GDF6 in promoting endothelial vascular integrity in vivo in zebrafish and in cultured human umbilical vein endothelial cells in vitro. We report that GDF6 promotes vascular integrity by counteracting vascular endothelial growth factor activity. GDF6-deficient endothelium has increased vascular endothelial growth factor signaling, increased vascular endothelial-cadherin Y658 phosphorylation, vascular endothelial-cadherin delocalization from cell-cell interfaces, and weakened endothelial cell adherence junctions that become prone to vascular leak. CONCLUSIONS: Our results suggest that GDF6 promotes vascular stabilization by restraining vascular endothelial growth factor signaling. Understanding how GDF6 affects vascular integrity may help to provide insights into hemorrhage and associated vascular pathologies in humans.


Subject(s)
Capillary Permeability , Embryo, Nonmammalian/blood supply , Endothelial Cells/metabolism , Growth Differentiation Factor 6/metabolism , Vascular Endothelial Growth Factor A/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Cells, Cultured , Gene Expression Regulation, Developmental , Growth Differentiation Factor 6/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mitogen-Activated Protein Kinase 3/metabolism , Neovascularization, Physiologic , Phosphorylation , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
14.
Development ; 143(17): 3182-94, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27578181

ABSTRACT

The receptor tyrosine kinase Ror2 is a major Wnt receptor that activates ß-catenin-independent signaling and plays a conserved role in the regulation of convergent extension movements and planar cell polarity in vertebrates. Mutations in the ROR2 gene cause recessive Robinow syndrome in humans, a short-limbed dwarfism associated with craniofacial malformations. Here, we show that Ror2 is required for local upregulation of gdf6 at the neural plate border in Xenopus embryos. Ror2 morphant embryos fail to upregulate neural plate border genes and show defects in the induction of neural crest cell fate. These embryos lack the spatially restricted activation of BMP signaling at the neural plate border at early neurula stages, which is required for neural crest induction. Ror2-dependent planar cell polarity signaling is required in the dorsolateral marginal zone during gastrulation indirectly to upregulate the BMP ligand Gdf6 at the neural plate border and Gdf6 is sufficient to rescue neural plate border specification in Ror2 morphant embryos. Thereby, Ror2 links Wnt/planar cell polarity signaling to BMP signaling in neural plate border specification and neural crest induction.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Growth Differentiation Factor 6/metabolism , Neural Plate/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Xenopus laevis/metabolism , Animals , Bone Morphogenetic Proteins/genetics , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Growth Differentiation Factor 6/genetics , Neural Crest/cytology , Neural Crest/embryology , Neural Crest/metabolism , Neural Plate/cytology , Neural Plate/embryology , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Xenopus laevis/embryology
15.
Aging (Albany NY) ; 8(6): 1259-75, 2016 06.
Article in English | MEDLINE | ID: mdl-27311402

ABSTRACT

The senescence-associated secretory phenotype (SASP) has attracted attention as a mechanism that connects cellular senescence to tissue dysfunction, and specific SASP factors have been identified as systemic pro-aging factors. However, little is known about the age-dependent changes in the secretory properties of stem cells. Young, but not old, mesenchymal stem/stromal cells (MSCs) are a well-known source of critical regenerative factors, but the identity of these factors remains elusive. In this study, we identified growth differentiation factor 6 (Gdf6; also known as Bmp13 and CDMP-2) as a regenerative factor secreted from young MSCs. The expression of specific secretory factors, including Gdf6, was regulated by the microRNA (miRNA) miR-17, whose expression declined with age. Upregulation of Gdf6 restored the osteogenic capacity of old MSCs in vitro and exerted positive effects in vivo on aging-associated pathologies such as reduced lymphopoiesis, insufficient muscle repair, reduced numbers of neural progenitors in the brain, and chronic inflammation. Our results suggest that manipulation of miRNA could enable control of the SASP, and that regenerative factors derived from certain types of young cells could be used to treat geriatric diseases.


Subject(s)
Cellular Senescence/physiology , Growth Differentiation Factor 6/metabolism , Mesenchymal Stem Cells/metabolism , Osteogenesis/physiology , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Cells, Cultured , Mice , MicroRNAs/metabolism , Up-Regulation
16.
Biotechnol Lett ; 38(5): 885-92, 2016 May.
Article in English | MEDLINE | ID: mdl-26956234

ABSTRACT

OBJECTIVES: To examine whether an engineered tendon matrix (ETM) environment and growth and differentiation factor-6 (GDF-6) have synergistic effects on the tenogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and the quality of tendon repair. RESULTS: ETM and GDF-6 promote tenogenic differentiation of BMSCs in vitro. Implantation of GDF-6-incorporated ETM containing BMSCs into a tendon injury model significantly improved the histological and mechanical properties of the repaired tendon. CONCLUSIONS: GDF-6-incorporated ETM containing BMSCs represents a promising strategy for tendon injury repair.


Subject(s)
Cell Differentiation , Extracellular Matrix/metabolism , Growth Differentiation Factor 6/metabolism , Mesenchymal Stem Cells/physiology , Regeneration , Tendons/physiology , Animals , Organ Culture Techniques , Rabbits , Rats
17.
Cell ; 164(1-2): 45-56, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26774823

ABSTRACT

Changes in bone size and shape are defining features of many vertebrates. Here we use genetic crosses and comparative genomics to identify specific regulatory DNA alterations controlling skeletal evolution. Armor bone-size differences in sticklebacks map to a major effect locus overlapping BMP family member GDF6. Freshwater fish express more GDF6 due in part to a transposon insertion, and transgenic overexpression of GDF6 phenocopies evolutionary changes in armor-plate size. The human GDF6 locus also has undergone distinctive regulatory evolution, including complete loss of an enhancer that is otherwise highly conserved between chimps and other mammals. Functional tests show that the ancestral enhancer drives expression in hindlimbs but not forelimbs, in locations that have been specifically modified during the human transition to bipedalism. Both gain and loss of regulatory elements can localize BMP changes to specific anatomical locations, providing a flexible regulatory basis for evolving species-specific changes in skeletal form.


Subject(s)
Biological Evolution , Evolution, Molecular , Growth Differentiation Factor 6/genetics , Skeleton/physiology , Vertebrates/genetics , Adaptation, Physiological , Animals , Enhancer Elements, Genetic , Fish Proteins/genetics , Fish Proteins/metabolism , Fresh Water , Growth Differentiation Factor 6/metabolism , Humans , Quantitative Trait Loci , Seawater , Skeleton/anatomy & histology , Smegmamorpha/genetics , Smegmamorpha/physiology , Species Specificity , Vertebrates/classification , Vertebrates/growth & development , Vertebrates/metabolism
18.
J Bone Miner Res ; 31(4): 882-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26643732

ABSTRACT

Growth and differentiation factors (GDFs) are secreted signaling molecules within the BMP family that have critical roles in joint morphogenesis during skeletal development in mice and humans. Using genetic data obtained from a six-generation Chinese family, we identified a missense variant in GDF6 (NP_001001557.1; p.Y444N) that fully segregates with a novel autosomal dominant synostoses (SYNS) phenotype, which we designate as SYNS4. Affected individuals display bilateral wrist and ankle deformities at birth and progressive conductive deafness after age 40 years. We find that the Y444N variant affects a highly conserved residue of GDF6 in a region critical for binding of GDF6 to its receptor(s) and to the BMP antagonist NOG, and show that this mutant GDF6 is a more potent stimulator of the canonical BMP signaling pathway compared with wild-type GDF6. Further, we determine that the enhanced BMP activity exhibited by mutant GDF6 is attributable to resistance to NOG-mediated antagonism. Collectively, our findings indicate that increased BMP signaling owing to a GDF6 gain-of-function mutation is responsible for loss of joint formation and profound functional impairment in patients with SYNS4. More broadly, our study highlights the delicate balance of BMP signaling required for proper joint morphogenesis and reinforces the critical role of BMP signaling in skeletal development.


Subject(s)
Bone Morphogenetic Proteins , Carpal Bones/abnormalities , Carrier Proteins , Foot Deformities, Congenital , Growth Differentiation Factor 6 , Hand Deformities, Congenital , Mutation, Missense , Signal Transduction/genetics , Stapes/abnormalities , Synostosis , Tarsal Bones/abnormalities , Amino Acid Substitution , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Carpal Bones/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Foot Deformities, Congenital/genetics , Foot Deformities, Congenital/metabolism , Growth Differentiation Factor 6/genetics , Growth Differentiation Factor 6/metabolism , Hand Deformities, Congenital/genetics , Hand Deformities, Congenital/metabolism , Humans , Mice , Stapes/metabolism , Synostosis/genetics , Synostosis/metabolism , Tarsal Bones/metabolism
19.
J Orthop Res ; 34(2): 279-89, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26184900

ABSTRACT

During embryogenesis vertebral segmentation is initiated by sclerotomal cell migration and condensation around the notochord, forming anlagen of vertebral bodies and intervertebral discs. The factors that govern the segmentation are not clear. Previous research demonstrated that mutations in growth differentiation factor 6 resulted in congenital vertebral fusion, suggesting this factor plays a role in development of vertebral column. In this study, we detected expression and localization of growth differentiation factor 6 in human fetal spinal column, especially in the period of early ossification of vertebrae and the developing intervertebral discs. The extracellular matrix proteins were also examined. Results showed that high levels of growth differentiation factor 6 were expressed in the nucleus pulposus of intervertebral discs and the hypertrophic chondrocytes adjacent to the ossification centre in vertebral bodies, where strong expression of proteoglycan and collagens was also detected. As fetal age increased, the expression of growth differentiation factor 6 was decreased correspondingly with the progress of ossification in vertebral bodies and restricted to cartilaginous regions. This expression pattern and the genetic link to vertebral fusion suggest that growth differentiation factor 6 may play an important role in suppression of ossification to ensure proper vertebral segmentation during spinal development.


Subject(s)
Fetal Development , Growth Differentiation Factor 6/metabolism , Spine/embryology , Cartilage/metabolism , Collagen/metabolism , Fetus/metabolism , Humans , Osteogenesis , Proteoglycans/metabolism , Spine/metabolism
20.
J Orthop Res ; 33(12): 1769-75, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26134557

ABSTRACT

Our laboratory has demonstrated that bone morphogenetic protein 13 prevented the effects of annular injury in an ovine model, maintaining intervertebral disc height, cell numbers and increasing extracellular matrix production compared to degenerated controls. The present study sought to examine the molecular effects of bone morphogenetic protein 13 on human degenerated disc cells and localize its expression in both human degenerate and scoliotic disc tissue. Effect of bone morphogenetic protein 13 on human derived nucleus pulposus, annulus fibrosus and endplate cells cultured in alginate beads was evaluated by changes in proteoglycan and collagen content. Migratory potential of disc cells towards bone morphogenetic protein 13 was also examined. Bone morphogenetic protein 13 induced significant proteoglycan accumulation in nucleus (18%), annulus (21%) and endplate (23%) cells cultured in alginate beads (p<0.05) compared to controls. Further bone morphogenetic protein 13 increased collagen I and II protein expression in nucleus and endplate cells. Nucleus cells displayed a significant chemotactic response towards bone morphogenetic protein 13. The endogenous expression of bone morphogenetic protein 13 in degenerate disc tissue was not different to scoliotic disc. Bone morphogenetic protein 13 has the potential to enhance extracellular matrix accumulation and induce cell migration in certain disc cells.


Subject(s)
Gene Expression Regulation , Growth Differentiation Factor 6/metabolism , Intervertebral Disc/cytology , Intervertebral Disc/metabolism , Adult , Alginates/chemistry , Cell Culture Techniques , Cell Movement , Chemotaxis , Female , Gene Expression Profiling , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Humans , Intervertebral Disc Degeneration/pathology , Male , Middle Aged , Proteoglycans/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...