Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Nat Commun ; 12(1): 6891, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34824272

ABSTRACT

Skeletal deformities are typical AD-HIES manifestations, which are mainly caused by heterozygous and loss-of-function mutations in Signal transducer and activator of transcription 3 (STAT3). However, the mechanism is still unclear and the treatment strategy is limited. Herein, we reported that the mice with Stat3 deletion in osteoblasts, but not in osteoclasts, induced AD-HIES-like skeletal defects, including craniofacial malformation, osteoporosis, and spontaneous bone fracture. Mechanistic analyses revealed that STAT3 in cooperation with Msh homeobox 1(MSX1) drove osteoblast differentiation by promoting Distal-less homeobox 5(Dlx5) transcription. Furthermore, pharmacological activation of STAT3 partially rescued skeletal deformities in heterozygous knockout mice, while inhibition of STAT3 aggravated bone loss. Taken together, these data show that STAT3 is critical for modulating skeletal development and maintaining bone homeostasis through STAT3-indcued osteogenesis and suggest it may be a potential target for treatments.


Subject(s)
Osteogenesis/genetics , STAT3 Transcription Factor/metabolism , Animals , Bone Development/genetics , Bone Remodeling/genetics , Cell Differentiation/drug effects , Homeodomain Proteins/genetics , Homeostasis/drug effects , Homeostasis/genetics , MSX1 Transcription Factor/genetics , MSX1 Transcription Factor/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Musculoskeletal Abnormalities/drug therapy , Musculoskeletal Abnormalities/genetics , Musculoskeletal Abnormalities/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/drug effects , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Signal Transduction , Transcription, Genetic
2.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Article in English | MEDLINE | ID: mdl-34548398

ABSTRACT

Skeletal ciliopathies (e.g., Jeune syndrome, short rib polydactyly syndrome, and Sensenbrenner syndrome) are frequently associated with nephronophthisis-like cystic kidney disease and other organ manifestations. Despite recent progress in genetic mapping of causative loci, a common molecular mechanism of cartilage defects and cystic kidneys has remained elusive. Targeting two ciliary chondrodysplasia loci (ift80 and ift172) by CRISPR/Cas9 mutagenesis, we established models for skeletal ciliopathies in Xenopus tropicalis Froglets exhibited severe limb deformities, polydactyly, and cystic kidneys, closely matching the phenotype of affected patients. A data mining-based in silico screen found ttc30a to be related to known skeletal ciliopathy genes. CRISPR/Cas9 targeting replicated limb malformations and renal cysts identical to the models of established disease genes. Loss of Ttc30a impaired embryonic renal excretion and ciliogenesis because of altered posttranslational tubulin acetylation, glycylation, and defective axoneme compartmentalization. Ttc30a/b transcripts are enriched in chondrocytes and osteocytes of single-cell RNA-sequenced embryonic mouse limbs. We identify TTC30A/B as an essential node in the network of ciliary chondrodysplasia and nephronophthisis-like disease proteins and suggest that tubulin modifications and cilia segmentation contribute to skeletal and renal ciliopathy manifestations of ciliopathies in a cell type-specific manner. These findings have implications for potential therapeutic strategies.


Subject(s)
Bone and Bones/abnormalities , Ciliopathies/pathology , Craniosynostoses/pathology , Cytoskeletal Proteins/metabolism , Ectodermal Dysplasia/pathology , Embryo, Nonmammalian/pathology , Musculoskeletal Abnormalities/pathology , Polycystic Kidney Diseases/pathology , Tubulin/chemistry , Animals , Bone and Bones/metabolism , Bone and Bones/pathology , Ciliopathies/genetics , Ciliopathies/metabolism , Craniosynostoses/genetics , Craniosynostoses/metabolism , Cytoskeletal Proteins/genetics , Disease Models, Animal , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/metabolism , Embryo, Nonmammalian/metabolism , Musculoskeletal Abnormalities/genetics , Musculoskeletal Abnormalities/metabolism , Phenotype , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/metabolism , Tubulin/metabolism , Xenopus laevis
3.
Int J Mol Sci ; 22(13)2021 Jun 22.
Article in English | MEDLINE | ID: mdl-34206401

ABSTRACT

Wingless-type MMTV integration site family, member 16 (wnt16), is a wnt ligand that participates in the regulation of vertebrate skeletal development. Studies have shown that wnt16 can regulate bone metabolism, but its molecular mechanism remains largely undefined. We obtained the wnt16-/- zebrafish model using the CRISPR-Cas9-mediated gene knockout screen with 11 bp deletion in wnt16, which led to the premature termination of amino acid translation and significantly reduced wnt16 expression, thus obtaining the wnt16-/- zebrafish model. The expression of wnt16 in bone-related parts was detected via in situ hybridization. The head, spine, and tail exhibited significant deformities, and the bone mineral density and trabecular bone decreased in wnt16-/- using light microscopy and micro-CT analysis. RNA sequencing was performed to explore the differentially expressed genes (DEGs). Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis found that the down-regulated DEGs are mainly concentrated in mTOR, FoxO, and VEGF pathways. Protein-protein interaction (PPI) network analysis was performed with the detected DEGs. Eight down-regulated DEGs including akt1, bnip4, ptena, vegfaa, twsg1b, prkab1a, prkab1b, and pla2g4f.2 were validated by qRT-PCR and the results were consistent with the RNA-seq data. Overall, our work provides key insights into the influence of wnt16 gene on skeletal development.


Subject(s)
Bone and Bones/abnormalities , Musculoskeletal Abnormalities/genetics , Musculoskeletal Abnormalities/metabolism , Osteogenesis/genetics , Wnt Proteins/deficiency , Zebrafish Proteins/deficiency , Zebrafish/genetics , Animals , Animals, Genetically Modified , Computational Biology/methods , Disease Models, Animal , Gene Expression Profiling , Gene Knockout Techniques , Gene Ontology , Molecular Sequence Annotation , Musculoskeletal Abnormalities/diagnosis , Phenotype , Transcriptome , Wnt Proteins/chemistry , Wnt Proteins/metabolism , Zebrafish Proteins/chemistry , Zebrafish Proteins/metabolism
4.
Am J Hum Genet ; 108(9): 1669-1691, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34314705

ABSTRACT

Transportin-2 (TNPO2) mediates multiple pathways including non-classical nucleocytoplasmic shuttling of >60 cargoes, such as developmental and neuronal proteins. We identified 15 individuals carrying de novo coding variants in TNPO2 who presented with global developmental delay (GDD), dysmorphic features, ophthalmologic abnormalities, and neurological features. To assess the nature of these variants, functional studies were performed in Drosophila. We found that fly dTnpo (orthologous to TNPO2) is expressed in a subset of neurons. dTnpo is critical for neuronal maintenance and function as downregulating dTnpo in mature neurons using RNAi disrupts neuronal activity and survival. Altering the activity and expression of dTnpo using mutant alleles or RNAi causes developmental defects, including eye and wing deformities and lethality. These effects are dosage dependent as more severe phenotypes are associated with stronger dTnpo loss. Interestingly, similar phenotypes are observed with dTnpo upregulation and ectopic expression of TNPO2, showing that loss and gain of Transportin activity causes developmental defects. Further, proband-associated variants can cause more or less severe developmental abnormalities compared to wild-type TNPO2 when ectopically expressed. The impact of the variants tested seems to correlate with their position within the protein. Specifically, those that fall within the RAN binding domain cause more severe toxicity and those in the acidic loop are less toxic. Variants within the cargo binding domain show tissue-dependent effects. In summary, dTnpo is an essential gene in flies during development and in neurons. Further, proband-associated de novo variants within TNPO2 disrupt the function of the encoded protein. Hence, TNPO2 variants are causative for neurodevelopmental abnormalities.


Subject(s)
Developmental Disabilities/genetics , Drosophila Proteins/genetics , Eye Diseases, Hereditary/genetics , Intellectual Disability/genetics , Karyopherins/genetics , Musculoskeletal Abnormalities/genetics , beta Karyopherins/genetics , ran GTP-Binding Protein/genetics , Alleles , Amino Acid Sequence , Animals , Developmental Disabilities/metabolism , Developmental Disabilities/pathology , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Eye Diseases, Hereditary/metabolism , Eye Diseases, Hereditary/pathology , Female , Gene Dosage , Gene Expression Regulation, Developmental , Genome, Human , Humans , Infant , Infant, Newborn , Intellectual Disability/metabolism , Intellectual Disability/pathology , Karyopherins/antagonists & inhibitors , Karyopherins/metabolism , Male , Musculoskeletal Abnormalities/metabolism , Musculoskeletal Abnormalities/pathology , Mutation , Neurons/metabolism , Neurons/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Whole Genome Sequencing , beta Karyopherins/metabolism , ran GTP-Binding Protein/metabolism
5.
PLoS Genet ; 17(6): e1009605, 2021 06.
Article in English | MEDLINE | ID: mdl-34081701

ABSTRACT

Homozygous mutation of the Csf1r locus (Csf1rko) in mice, rats and humans leads to multiple postnatal developmental abnormalities. To enable analysis of the mechanisms underlying the phenotypic impacts of Csf1r mutation, we bred a rat Csf1rko allele to the inbred dark agouti (DA) genetic background and to a Csf1r-mApple reporter transgene. The Csf1rko led to almost complete loss of embryonic macrophages and ablation of most adult tissue macrophage populations. We extended previous analysis of the Csf1rko phenotype to early postnatal development to reveal impacts on musculoskeletal development and proliferation and morphogenesis in multiple organs. Expression profiling of 3-week old wild-type (WT) and Csf1rko livers identified 2760 differentially expressed genes associated with the loss of macrophages, severe hypoplasia, delayed hepatocyte maturation, disrupted lipid metabolism and the IGF1/IGF binding protein system. Older Csf1rko rats developed severe hepatic steatosis. Consistent with the developmental delay in the liver Csf1rko rats had greatly-reduced circulating IGF1. Transfer of WT bone marrow (BM) cells at weaning without conditioning repopulated resident macrophages in all organs, including microglia in the brain, and reversed the mutant phenotypes enabling long term survival and fertility. WT BM transfer restored osteoclasts, eliminated osteopetrosis, restored bone marrow cellularity and architecture and reversed granulocytosis and B cell deficiency. Csf1rko rats had an elevated circulating CSF1 concentration which was rapidly reduced to WT levels following BM transfer. However, CD43hi non-classical monocytes, absent in the Csf1rko, were not rescued and bone marrow progenitors remained unresponsive to CSF1. The results demonstrate that the Csf1rko phenotype is autonomous to BM-derived cells and indicate that BM contains a progenitor of tissue macrophages distinct from hematopoietic stem cells. The model provides a unique system in which to define the pathways of development of resident tissue macrophages and their local and systemic roles in growth and organ maturation.


Subject(s)
Fatty Liver/genetics , Macrophages/metabolism , Musculoskeletal Abnormalities/genetics , Musculoskeletal Development/genetics , Osteopetrosis/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Bone Marrow Transplantation , Disease Models, Animal , Embryo, Mammalian , Fatty Liver/metabolism , Fatty Liver/pathology , Fatty Liver/therapy , Female , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Genes, Reporter , Humans , Insulin-Like Growth Factor Binding Proteins/deficiency , Insulin-Like Growth Factor Binding Proteins/genetics , Insulin-Like Growth Factor I/deficiency , Insulin-Like Growth Factor I/genetics , Lipid Metabolism , Liver/metabolism , Liver/pathology , Macrophages/pathology , Male , Musculoskeletal Abnormalities/metabolism , Musculoskeletal Abnormalities/pathology , Musculoskeletal Abnormalities/therapy , Osteopetrosis/metabolism , Osteopetrosis/pathology , Osteopetrosis/therapy , Rats , Rats, Transgenic , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/deficiency
6.
Am J Hum Genet ; 107(4): 763-777, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32937143

ABSTRACT

Distal hereditary motor neuropathies (HMNs) and axonal Charcot-Marie-Tooth neuropathy (CMT2) are clinically and genetically heterogeneous diseases characterized primarily by motor neuron degeneration and distal weakness. The genetic cause for about half of the individuals affected by HMN/CMT2 remains unknown. Here, we report the identification of pathogenic variants in GBF1 (Golgi brefeldin A-resistant guanine nucleotide exchange factor 1) in four unrelated families with individuals affected by sporadic or dominant HMN/CMT2. Genomic sequencing analyses in seven affected individuals uncovered four distinct heterozygous GBF1 variants, two of which occurred de novo. Other known HMN/CMT2-implicated genes were excluded. Affected individuals show HMN/CMT2 with slowly progressive distal muscle weakness and musculoskeletal deformities. Electrophysiological studies confirmed axonal damage with chronic neurogenic changes. Three individuals had additional distal sensory loss. GBF1 encodes a guanine-nucleotide exchange factor that facilitates the activation of members of the ARF (ADP-ribosylation factor) family of small GTPases. GBF1 is mainly involved in the formation of coatomer protein complex (COPI) vesicles, maintenance and function of the Golgi apparatus, and mitochondria migration and positioning. We demonstrate that GBF1 is present in mouse spinal cord and muscle tissues and is particularly abundant in neuropathologically relevant sites, such as the motor neuron and the growth cone. Consistent with the described role of GBF1 in Golgi function and maintenance, we observed marked increase in Golgi fragmentation in primary fibroblasts derived from all affected individuals in this study. Our results not only reinforce the existing link between Golgi fragmentation and neurodegeneration but also demonstrate that pathogenic variants in GBF1 are associated with HMN/CMT2.


Subject(s)
Axons/metabolism , Charcot-Marie-Tooth Disease/genetics , Guanine Nucleotide Exchange Factors/genetics , Muscle Weakness/genetics , Muscular Atrophy, Spinal/genetics , Musculoskeletal Abnormalities/genetics , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Animals , Axons/pathology , COP-Coated Vesicles/metabolism , COP-Coated Vesicles/pathology , Charcot-Marie-Tooth Disease/diagnosis , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression , Golgi Apparatus/metabolism , Golgi Apparatus/pathology , Guanine Nucleotide Exchange Factors/metabolism , Heterozygote , Humans , Male , Mice , Middle Aged , Mitochondria/metabolism , Mitochondria/pathology , Motor Neurons/metabolism , Motor Neurons/pathology , Muscle Weakness/diagnosis , Muscle Weakness/metabolism , Muscle Weakness/pathology , Muscular Atrophy, Spinal/diagnosis , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Musculoskeletal Abnormalities/diagnosis , Musculoskeletal Abnormalities/metabolism , Musculoskeletal Abnormalities/pathology , Mutation , Pedigree , Primary Cell Culture , Spinal Cord/abnormalities , Spinal Cord/metabolism
7.
Biochem Biophys Res Commun ; 530(3): 520-526, 2020 09 24.
Article in English | MEDLINE | ID: mdl-32620236

ABSTRACT

PIK3CA-related overgrowth spectrum is caused by mosaicism mutations in the PIK3CA gene. These mutations, which are also observed in various types of cancer, lead to a constitutive activation of the PI3K/AKT/mTOR pathway, increasing cell proliferation. Heat shock transcription factor 1 (HSF1) is the major stress-responsive transcription factor. Recent findings indicate that AKT phosphorylates and activates HSF1 independently of heat-shock in breast cancer cells. Here, we aimed to investigate the role of HSF1 in PIK3CA-related overgrowth spectrum. We observed a higher rate of proliferation and increased phosphorylation of AKT and p70S6K in mutant fibroblasts than in control cells. We also found elevated phosphorylation and activation of HSF1, which is directly correlated to AKT activation. Specific AKT inhibitors inhibit HSF1 phosphorylation as well as HSF1-dependent gene transcription. Finally, we demonstrated that targeting HSF1 with specific inhibitors reduced the proliferation of mutant cells. As there is currently no curative treatment for PIK3CA-related overgrowth spectrum, our results identify HSF1 as a new potential therapeutic target.


Subject(s)
Cell Proliferation/drug effects , Class I Phosphatidylinositol 3-Kinases/metabolism , Drug Discovery , Heat Shock Transcription Factors/antagonists & inhibitors , Lipoma/metabolism , Musculoskeletal Abnormalities/metabolism , Nevus/metabolism , Vascular Malformations/metabolism , Cells, Cultured , Class I Phosphatidylinositol 3-Kinases/genetics , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Heat Shock Transcription Factors/metabolism , Humans , Lipoma/drug therapy , Lipoma/genetics , Lipoma/pathology , Molecular Targeted Therapy , Musculoskeletal Abnormalities/drug therapy , Musculoskeletal Abnormalities/genetics , Musculoskeletal Abnormalities/pathology , Mutation , Nevus/drug therapy , Nevus/genetics , Nevus/pathology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Vascular Malformations/drug therapy , Vascular Malformations/genetics , Vascular Malformations/pathology
8.
Nat Commun ; 11(1): 3168, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32576830

ABSTRACT

In humans, mutations in the PIEZO2 gene, which encodes for a mechanosensitive ion channel, were found to result in skeletal abnormalities including scoliosis and hip dysplasia. Here, we show in mice that loss of Piezo2 expression in the proprioceptive system recapitulates several human skeletal abnormalities. While loss of Piezo2 in chondrogenic or osteogenic lineages does not lead to human-like skeletal abnormalities, its loss in proprioceptive neurons leads to spine malalignment and hip dysplasia. To validate the non-autonomous role of proprioception in hip joint morphogenesis, we studied this process in mice mutant for proprioceptive system regulators Runx3 or Egr3. Loss of Runx3 in the peripheral nervous system, but not in skeletal lineages, leads to similar joint abnormalities, as does Egr3 loss of function. These findings expand the range of known regulatory roles of the proprioception system on the skeleton and provide a central component of the underlying molecular mechanism, namely Piezo2.


Subject(s)
Ion Channels/metabolism , Musculoskeletal Abnormalities/metabolism , Musculoskeletal System/metabolism , Neurons/metabolism , Proprioception/physiology , Abnormalities, Multiple , Animals , Bone Remodeling , Core Binding Factor Alpha 3 Subunit/metabolism , Disease Models, Animal , Early Growth Response Protein 3/metabolism , Genetic Predisposition to Disease/genetics , Hip Dislocation/genetics , Hip Dislocation/metabolism , Hip Dislocation/pathology , Hip Joint/anatomy & histology , Hip Joint/metabolism , Hip Joint/pathology , Ion Channels/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Musculoskeletal Abnormalities/genetics , Musculoskeletal Abnormalities/pathology , Musculoskeletal System/pathology , Scoliosis
9.
Article in English | MEDLINE | ID: mdl-32328030

ABSTRACT

Skeletal dysplasias are a diverse group of heritable diseases affecting bone and cartilage growth. Throughout the years, the molecular defect underlying many of the diseases has been identified. These identifications led to novel insights in the mechanisms regulating bone and cartilage growth and homeostasis. One of the pathways that is clearly important during skeletal development and bone homeostasis is the Wingless and int-1 (WNT) signaling pathway. So far, three different WNT signaling pathways have been described, which are all activated by binding of the WNT ligands to the Frizzled (FZD) receptors. In this review, we discuss the skeletal disorders that are included in the latest nosology of skeletal disorders and that are caused by genetic defects involving the WNT signaling pathway. The number of skeletal disorders caused by defects in WNT signaling genes and the clinical phenotype associated with these disorders illustrate the importance of the WNT signaling pathway during skeletal development as well as later on in life to maintain bone mass. The knowledge gained through the identification of the genes underlying these monogenic conditions is used for the identification of novel therapeutic targets. For example, the genes underlying disorders with altered bone mass are all involved in the canonical WNT signaling pathway. Consequently, targeting this pathway is one of the major strategies to increase bone mass in patients with osteoporosis. In addition to increasing the insights in the pathways regulating skeletal development and bone homeostasis, knowledge of rare skeletal dysplasias can also be used to predict possible adverse effects of these novel drug targets. Therefore, this review gives an overview of the skeletal and extra-skeletal phenotype of the different skeletal disorders linked to the WNT signaling pathway.


Subject(s)
Bone Diseases/genetics , Bone and Bones/physiology , Musculoskeletal Abnormalities/genetics , Wnt Signaling Pathway/physiology , Animals , Bone Diseases/metabolism , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Humans , Musculoskeletal Abnormalities/metabolism , Wnt Signaling Pathway/genetics
11.
J Hazard Mater ; 387: 121720, 2020 04 05.
Article in English | MEDLINE | ID: mdl-31812480

ABSTRACT

Excess selenium entering the aquatic environment from anthropogenic activities has been associated with developmental abnormalities in fish including skeletal deformities of the head and spine. However, mechanisms of this developmental toxicity have not been well-characterized. In this study, Japanese medaka (Oryzias latipes) embryos were exposed to seleno-l-methionine (Se-Met) in a range of concentrations. Gene expression was evaluated for sex-determining region Y (SRY)-related box (Sox9a and Sox9b), runt-related transcription factor 2 (Runx2), and melatonin receptor (Mtr). Alterations in the length of Meckel's cartilage, tail curvature, and decreased calcification were observed in skeletal stains at 10- and 22-days post-fertilization (dpf). Embryonic exposure of Osterix-mCherry transgenic medaka resulted in fewer teeth. Sox9a and Sox9b were up-regulated, while Runx2 and Mtr were down-regulated by Se-Met prior to hatch. Whole mount in situ hybridization (WISH) localized gene expression to areas observed to be affected in vivo. In addition, Se-Met exposures of a Mtr morpholino (Mtr-MO) as well as Luzindole exposed embryos developed similar skeletal malformations, supporting involvement of Mtr. These findings demonstrate that Se-Met modulates expression of key genes involved in chondrogenic differentiation and bone formation during development.


Subject(s)
Cell Differentiation/drug effects , Chondrocytes/drug effects , Embryonic Development/drug effects , Musculoskeletal Abnormalities/chemically induced , Selenomethionine/toxicity , Water Pollutants, Chemical/toxicity , Animals , Core Binding Factor Alpha 1 Subunit/metabolism , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Fish Proteins/metabolism , Gene Expression/drug effects , Musculoskeletal Abnormalities/metabolism , Oryzias/embryology , Receptors, Melatonin/metabolism , SOX9 Transcription Factor/metabolism
12.
Eur Spine J ; 28(9): 1977-1986, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31079250

ABSTRACT

PURPOSE: To determine the relationship between the bone formation-related functions of GPR126 and the structural asymmetry of spine in adolescent idiopathic scoliosis (AIS). METHODS: Vertebral body samples were obtained from 51 AIS patients during spinal surgery between October 2014 and November 2017, and the expression pattern of GPR126 in the convex/concave sides of AIS spine was identified by RT-qPCR. Next, we explored the bone formation-related functions of GPR126 by knocking down and overexpressing GPR126 in human mesenchymal stem cells (hMSC) and further performing osteogenic differentiation. We also applied overexpression of N-terminal fragments derived from GPR126 (GPR126-NTFs) and osteogenic differentiation experiments to determine the functional part of GPR126 in skeletal development. RESULTS: We provided evidence that GPR126 showed a marked convex/concave asymmetric expression in the spine of AIS. Further RNA detection found that exon6-included transcripts of GPR126 (GPR126-exon6in) were significantly higher expressed in the convex side of AIS patients. Knocking down of GPR126 accelerated ossification of hMSCs during osteogenic differentiation, and overexpression of GPR126-exon6in delayed this process. Overexpression of GPR126-NTFs revealed that NTF is a functional fragment and exon6-included NTF (NTF-exon6in) delayed ossification of hMSCs. CONCLUSION: Our findings indicated that GPR126-NTFs play a role in skeletal development, and the inclusion/exclusion of exon6 may regulate the bone formation-related functions of GPR126. The convex/concave asymmetric expression of GPR126-exon6in may be an important factor in abnormal bone formation of AIS. These slides can be retrieved under Electronic Supplementary Material.


Subject(s)
Musculoskeletal Abnormalities/metabolism , Receptors, G-Protein-Coupled/biosynthesis , Scoliosis/metabolism , Adolescent , Cell Differentiation/physiology , Cells, Cultured , Child , Female , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Humans , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Musculoskeletal Abnormalities/genetics , Musculoskeletal Abnormalities/pathology , Osteogenesis/genetics , Osteogenesis/physiology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Scoliosis/genetics , Scoliosis/pathology , Scoliosis/surgery , Spine/pathology , Spine/physiopathology
13.
Birth Defects Res ; 111(5): 237-247, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30719872

ABSTRACT

BACKGROUND: The mutant chondrodysplasia (cho) is a cartilage-targeting disorder in C57BL mice that results in dwarfing and other malformations stemming from this collagenopathy. Clarke Fraser made the discovery of the mutation accidentally in the early 1960s during the thalidomide tragedy. METHODS: For this review we identified key research on cho as since its discovery. Relevant data were compiled to make a comprehensive review that details discoveries associated with the cho mutation, that describes the associated phenotypes and molecular mechanisms, and that provides a discussion surrounding its current clinical relevance. RESULTS: Mechanistically, cho acts by hindering chondrogenesis and endochondral bone formation. The phenotype results from a 1-nt deletion in the gene encoding the alpha 1 chain of type XI collagen. For more than half a century, researchers have studied the pathogenesis of the cho mutation in relation to a variety of mouse models of human birth defects and disease. These studies have resulted in several discoveries linking cho with such human disorders as dwarfism, tracheal stenosis, cleft palate, pulmonary hypoplasia, and osteoarthritis (OA). CONCLUSION: The study of cho has led to numerous advances in understanding human birth defects, congenital disorders, and adult human disease. The most recent studies have suggested a role for the TGF-Beta, HtrA1, Ddr2, and Mmp-13 pathway in the degradation of articular cartilage and the development of OA in cho/+ mice. We have shown that the anti-hypertension drug Losartan is a TGF-Beta blocker that could be used to treat OA in Stickler syndrome, and thereby rescue the WT phenotype.


Subject(s)
Osteochondrodysplasias/genetics , Osteochondrodysplasias/metabolism , Osteochondrodysplasias/physiopathology , Abnormalities, Multiple/metabolism , Animals , Cartilage, Articular , Collagen Type XI/genetics , Collagen Type XI/metabolism , Disease Models, Animal , Lung/abnormalities , Lung/metabolism , Lung Diseases/metabolism , Mice , Mice, Inbred C57BL , Musculoskeletal Abnormalities/metabolism , Mutation , Osteoarthritis , Phenotype
14.
Front Neuroendocrinol ; 52: 113-143, 2019 01.
Article in English | MEDLINE | ID: mdl-30448536

ABSTRACT

Individuals with acromegaloid physical appearance or tall stature may be referred to endocrinologists to exclude growth hormone (GH) excess. While some of these subjects could be healthy individuals with normal variants of growth or physical traits, others will have acromegaly or pituitary gigantism, which are, in general, straightforward diagnoses upon assessment of the GH/IGF-1 axis. However, some patients with physical features resembling acromegaly - usually affecting the face and extremities -, or gigantism - accelerated growth/tall stature - will have no abnormalities in the GH axis. This scenario is termed pseudoacromegaly, and its correct diagnosis can be challenging due to the rarity and variability of these conditions, as well as due to significant overlap in their characteristics. In this review we aim to provide a comprehensive overview of pseudoacromegaly conditions, highlighting their similarities and differences with acromegaly and pituitary gigantism, to aid physicians with the diagnosis of patients with pseudoacromegaly.


Subject(s)
Acromegaly/diagnosis , Chromosome Disorders/diagnosis , Diagnosis, Differential , Gigantism/diagnosis , Glucose Metabolism Disorders/diagnosis , Hypothyroidism/diagnosis , Lipodystrophy/diagnosis , Marfan Syndrome/diagnosis , Musculoskeletal Abnormalities/diagnosis , Osteochondrodysplasias/diagnosis , Acromegaly/metabolism , Chromosome Disorders/metabolism , Gigantism/metabolism , Glucose Metabolism Disorders/metabolism , Humans , Lipodystrophy/metabolism , Musculoskeletal Abnormalities/metabolism
15.
J Cell Physiol ; 234(1): 231-245, 2018 01.
Article in English | MEDLINE | ID: mdl-30076721

ABSTRACT

MiR-214 belongs to a family of microRNA (small, highly conserved noncoding RNA molecules) precursors that play a pivotal role in biological functions, such as cellular function, tissue development, tissue homeostasis, and pathogenesis of diseases. Recently, miR-214 emerged as a critical regulator of musculoskeletal metabolism. Specifically, miR-214 can mediate skeletal muscle myogenesis and vascular smooth muscle cell proliferation, migration, and differentiation. MiR-214 also modulates osteoblast function by targeting specific molecular pathways and the expression of various osteoblast-related genes; promotes osteoclast activity by targeting phosphatase and tensin homolog (Pten); and mediates osteoclast-osteoblast intercellular crosstalk via an exosomal miRNA paracrine mechanism. Importantly, dysregulation in miR-214 expression is associated with pathological bone conditions such as osteoporosis, osteosarcoma, multiple myeloma, and osteolytic bone metastasis of breast cancer. This review discusses the cellular targets of miR-214 in bone, the molecular mechanisms governing the activities of miR-214 in the musculoskeletal system, and the putative role of miR-214 in skeletal diseases. Understanding the biology of miR-214 could potentially lead to the development of miR-214 as a possible biomarker and a therapeutic target for musculoskeletal diseases.


Subject(s)
Bone Neoplasms/genetics , MicroRNAs/genetics , Muscle, Skeletal/metabolism , Musculoskeletal Abnormalities/genetics , Biomarkers, Tumor/genetics , Bone Neoplasms/pathology , Cell Differentiation/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Muscle, Skeletal/pathology , Musculoskeletal Abnormalities/metabolism , Musculoskeletal Abnormalities/pathology , Osteoblasts/metabolism , Osteoblasts/pathology , PTEN Phosphohydrolase/genetics
16.
Birth Defects Res ; 109(10): 791-804, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28509418

ABSTRACT

The most impressive phenotypic appearance of sirenomelia is the presence of a 180°-rotated, axially positioned, single lower limb. Associated gastrointestinal and genitourinary anomalies are almost always present. This rare anomaly is still the subject of ongoing controversies concerning its nosology, pathogenesis, and possible genetic etiology. Sirenomelia can be part of a syndromic continuum, overlapping with other complex conditions including caudal dysgenesis and VATER/VACTERL/VACTERL-H associations, which could all be part of a heterogeneous spectrum, and originate from an early defect in blastogenesis. It is imaginable that different "primary field defects," whether or not genetically based, induce a spectrum of caudal malformations. In the current study, we review the contemporary hypotheses and conceptual approaches regarding the etiology and pathogenesis of sirenomelia, especially in the context of concomitant conditions. To expand on the latter, we included the external and internal dysmorphology of one third trimester sirenomelic fetus from our anatomical museum collection, in which multiple concomitant but discordant anomalies were observed compared with classic sirenomelia, and was diagnosed as VACTERL-H association with sirenomelia. Birth Defects Research 109:791-804, 2017. © 2017 The Authors. Birth Defects Research Published by Wiley Periodicals, Inc.


Subject(s)
Ectromelia/metabolism , Ectromelia/physiopathology , Abnormalities, Multiple/pathology , Anal Canal/abnormalities , Anal Canal/metabolism , Anal Canal/physiopathology , Cardiovascular Abnormalities/metabolism , Cardiovascular Abnormalities/physiopathology , Digestive System Abnormalities/metabolism , Digestive System Abnormalities/physiopathology , Ectromelia/complications , Ectromelia/diagnosis , Esophagus/abnormalities , Esophagus/metabolism , Esophagus/physiopathology , Fetus/abnormalities , Genetic Diseases, X-Linked/metabolism , Genetic Diseases, X-Linked/physiopathology , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/physiopathology , Humans , Hydrocephalus/metabolism , Hydrocephalus/physiopathology , Kidney/abnormalities , Kidney/metabolism , Kidney/physiopathology , Limb Deformities, Congenital/metabolism , Limb Deformities, Congenital/physiopathology , Musculoskeletal Abnormalities/metabolism , Musculoskeletal Abnormalities/physiopathology , Spine/abnormalities , Spine/metabolism , Spine/physiopathology , Trachea/abnormalities , Trachea/metabolism , Trachea/physiopathology , Urogenital Abnormalities/etiology , Urogenital Abnormalities/physiopathology
17.
J Bone Miner Res ; 31(11): 1930-1942, 2016 11.
Article in English | MEDLINE | ID: mdl-27541483

ABSTRACT

Bruck syndrome (BS) is a disorder characterized by joint flexion contractures and skeletal dysplasia that shows strong clinical overlap with the brittle bone disease osteogenesis imperfecta (OI). BS is caused by biallelic mutations in either the FKBP10 or the PLOD2 gene. PLOD2 encodes the lysyl hydroxylase 2 (LH2) enzyme, which is responsible for the hydroxylation of lysine residues in fibrillar collagen telopeptides. This hydroxylation directs crosslinking of collagen fibrils in the extracellular matrix, which is necessary to provide stability and tensile integrity to the collagen fibrils. To further elucidate the function of LH2 in vertebrate skeletal development, we created a zebrafish model harboring a homozygous plod2 nonsense mutation resulting in reduced telopeptide hydroxylation and crosslinking of bone type I collagen. Adult plod2 mutants present with a shortened body axis and severe skeletal abnormalities with evidence of bone fragility and fractures. The vertebral column of plod2 mutants is short and scoliotic with compressed vertebrae that show excessive bone formation at the vertebral end plates, and increased tissue mineral density in the vertebral centra. The muscle fibers of mutant zebrafish have a reduced diameter near the horizontal myoseptum. The endomysium, a layer of connective tissue ensheathing the individual muscle fibers, is enlarged. Transmission electron microscopy of mutant vertebral bone shows type I collagen fibrils that are less organized with loss of the typical plywood-like structure. In conclusion, plod2 mutant zebrafish show molecular and tissue abnormalities in the musculoskeletal system that are concordant with clinical findings in BS patients. Therefore, the plod2 zebrafish mutant is a promising model for the elucidation of the underlying pathogenetic mechanisms leading to BS and the development of novel therapeutic avenues in this syndrome. © 2016 American Society for Bone and Mineral Research.


Subject(s)
Arthrogryposis/pathology , Collagen Type I/metabolism , Lysine/metabolism , Musculoskeletal Abnormalities/pathology , Osteogenesis Imperfecta/pathology , Peptides/metabolism , Zebrafish/metabolism , Amino Acid Sequence , Animals , Arthrogryposis/complications , Arthrogryposis/diagnostic imaging , Arthrogryposis/metabolism , Bone and Bones/abnormalities , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Calcification, Physiologic , Catalytic Domain , Codon, Nonsense/genetics , Conserved Sequence/genetics , Cross-Linking Reagents/metabolism , Evolution, Molecular , Hydroxylation , Larva/metabolism , Mass Spectrometry , Musculoskeletal Abnormalities/complications , Musculoskeletal Abnormalities/diagnostic imaging , Musculoskeletal Abnormalities/metabolism , Notochord/pathology , Osteogenesis Imperfecta/complications , Osteogenesis Imperfecta/diagnostic imaging , Osteogenesis Imperfecta/metabolism , Phenotype , X-Ray Microtomography , Zebrafish Proteins/genetics
18.
Matrix Biol ; 37: 102-11, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24997222

ABSTRACT

Thrombospondins (TSPs) are widely known as a family of five calcium-binding matricellular proteins. While these proteins belong to the same family, they are encoded by different genes, regulate different cellular functions and are localized to specific regions of the body. TSP-5 or Cartilage Oligomeric Matrix Protein (COMP) is the only TSP that has been associated with skeletal disorders in humans, including pseudoachondroplasia (PSACH) and multiple epiphyseal dysplasia (MED). The pentameric structure of COMP, the evidence that it interacts with multiple cellular proteins, and the recent reports of COMP acting as a 'lattice' to present growth factors to cells, inspired this review of COMP and its interacting partners. In our review, we have compiled the interactions of COMP with other proteins in the cartilage extracellular matrix and summarized their importance in maintaining the structural integrity of cartilage as well as in regulating cellular functions.


Subject(s)
Cartilage Oligomeric Matrix Protein/metabolism , Chondrogenesis/physiology , Extracellular Matrix Proteins/metabolism , Musculoskeletal Abnormalities/metabolism , Humans , Models, Biological , Musculoskeletal Abnormalities/genetics , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary
19.
BMC Res Notes ; 7: 195, 2014 Mar 29.
Article in English | MEDLINE | ID: mdl-24679067

ABSTRACT

BACKGROUND: Congenital anomalies or birth defects are among the leading causes of infant mortality and morbidity around the world. The impact of congenital anomalies is particularly severe in middle- and low-income countries where health care resources are limited. The prevalence of congenital anomalies varies in different parts of the world, which could reflect different aetiological factors in different geographical regions. METHODS: Between October 2012 and January 2013, a cross-sectional study was conducted involving young infants below 2 months of age, admitted at a university teaching hospital in Tanzania. Face-to-face interviews with parents/caretakers of young infants were carried out to collect socio-demographic and clinical information. Physical examinations were performed on all young infants. Echocardiography, X-ray, cranial as well as abdominal ultrasonographies were performed when indicated. RESULTS: Analysis of the data showed that among 445 young infants enrolled in the study, the prevalence of congenital anomalies was 29%, with the Central Nervous System (CNS) as the most commonly affected organ system. Maternal factors that were significantly associated with congenital anomalies included the lack of peri-conceptional use of folic acid (OR = 3.1; 95% CI = 1.4-6.7; p = 0.005), a maternal age of above 35 years (OR = 2.2; 95% CI = 1.1-4.3; p = 0.024) and an inadequate attendance to antenatal clinic (OR = 2.1; 95% CI = 1.4-3.3; p < 0.001). Infant factors that were significantly associated with congenital anomalies were female sex, a birth weight of 2.5 kg or more, singleton pregnancy and a birth order above 4. CONCLUSIONS: Due to the high prevalence of congenital anomalies observed in this particular context, the hospital should mobilize additional resources for an optimal and timely management of the patients with congenital anomalies. In this study, the proportion of women taking folic acid supplements during early pregnancy was very low. Efforts should be made to ensure that more women use folic acid during the peri-conceptional period, as the use of folic acid supplement has been linked by several authors to a reduced occurrence of some congenital anomalies.


Subject(s)
Folic Acid Deficiency/epidemiology , Musculoskeletal Abnormalities/epidemiology , Nervous System Malformations/epidemiology , Adult , Birth Weight , Cross-Sectional Studies , Dietary Supplements , Female , Folic Acid , Folic Acid Deficiency/metabolism , Humans , Infant , Infant, Newborn , Male , Maternal Age , Musculoskeletal Abnormalities/metabolism , Nervous System Malformations/metabolism , Pregnancy , Prenatal Care/organization & administration , Risk Factors , Tanzania/epidemiology
20.
Hum Genet ; 132(11): 1253-64, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23812741

ABSTRACT

All TGF-beta family members have a prodomain that is important for secretion. Lack of secretion of a TGF-beta family member GDF5 is known to underlie some skeletal abnormalities, such as brachydactyly type C that is characterized by a huge and unexplained phenotypic variability. To search for potential phenotypic modifiers regulating secretion of GDF5, we compared cells overexpressing wild type (Wt) GDF5 and GDF5 with a novel mutation in the prodomain identified in a large Pakistani family with Brachydactyly type C and mild Grebe type chondrodyslplasia (c527T>C; p.Leu176Pro). Initial in vitro expression studies revealed that the p.Leu176Pro mutant (Mut) GDF5 was not secreted outside the cells. We subsequently showed that GDF5 was capable of forming a complex with latent transforming growth factor binding proteins, LTBP1 and LTBP2. Furthermore, secretion of LTBP1 and LTBP2 was severely impaired in cells expressing the Mut-GDF5 compared to Wt-GDF5. Finally, we demonstrated that secretion of Wt-GDF5 was inhibited by the Mut-GDF5, but only when LTBP (LTBP1 or LTBP2) was co-expressed. Based on these findings, we suggest a novel model, where the dosage of secretory co-factors or stabilizing proteins like LTBP1 and LTBP2 in the microenvironment may affect the extent of GDF5 secretion and thereby function as modifiers in phenotypes caused by GDF5 mutations.


Subject(s)
Asian People/genetics , Brachydactyly/genetics , Growth Differentiation Factor 5/genetics , Musculoskeletal Abnormalities/genetics , Mutation, Missense , Osteochondrodysplasias/genetics , Amino Acid Sequence , Brachydactyly/physiopathology , Genotype , Growth Differentiation Factor 5/metabolism , HEK293 Cells , Humans , Immunoprecipitation , Latent TGF-beta Binding Proteins/genetics , Latent TGF-beta Binding Proteins/metabolism , Molecular Sequence Data , Musculoskeletal Abnormalities/metabolism , Osteochondrodysplasias/metabolism , Pakistan , Pedigree , Phenotype , Protein Conformation , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...