Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Am J Med Genet A ; 191(10): 2631-2639, 2023 10.
Article in English | MEDLINE | ID: mdl-37353357

ABSTRACT

Myopathic Ehlers-Danlos syndrome (mEDS) is a subtype of EDS that is caused by abnormalities in COL12A1. Up-to-date, 24 patients from 15 families with mEDS have been reported, with 14 families showing inheritance in an autosomal dominant manner and one family in an autosomal recessive manner. We encountered an additional patient with autosomal recessive mEDS. The patient is a 47-year-old Japanese man, born to consanguineous parents with no related features of mEDS. After birth, he presented with hypotonia, weak spontaneous movements, scoliosis, and torticollis. He had soft palms but no skin hyperextensibility or fragility. Progressive scoliosis, undescended testes, and muscular torticollis required surgery. During adulthood, he worked normally and had no physical concerns. Clinical exome analysis revealed a novel homozygous variant in COL12A1 (NM_004370.6:c.395-1G > A) at the splice acceptor site of exon 6, leading to in-frame skipping of exon 6. The patient was diagnosed with mEDS. The milder manifestations in the current patient compared with previously reported patients with mEDS might be related to the site of the variant. The variant is located in the genomic region encoding the first von Willebrand factor A domain, which affects only the long isoform of collagen XII, in contrast to the variants in previously reported mEDS patients that affected both the long and short isoforms. Further studies are needed to delineate comprehensive genotype-phenotype correlation of the disorder.


Subject(s)
Ehlers-Danlos Syndrome , Scoliosis , Torticollis , Humans , Male , Middle Aged , Collagen/genetics , Collagen Type XII/genetics , Ehlers-Danlos Syndrome/complications , Ehlers-Danlos Syndrome/diagnosis , Ehlers-Danlos Syndrome/genetics , Mutation , von Willebrand Factor/genetics
2.
Int J Mol Sci ; 24(21)2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37958789

ABSTRACT

Several methods have been developed to generate neurons from other cell types for performing regeneration therapy and in vitro studies of central nerve disease. Small molecules (SMs) can efficiently induce neuronal features in human and rodent fibroblasts without transgenes. Although canines have been used as a spontaneous disease model of human central nerve, efficient neuronal reprogramming method of canine cells have not been well established. We aimed to induce neuronal features in adult canine dermal fibroblasts (ACDFs) by SMs and assess the permanency of these changes. ACDFs treated with eight SMs developed a round-shaped cell body with branching processes and expressed neuronal proteins, including ßIII-tubulin, microtubule-associated protein 2 (MAP2), and neurofilament-medium. Transcriptome profiling revealed the upregulation of neuron-related genes, such as SNAP25 and GRIA4, and downregulation of fibroblast-related genes, such as COL12A1 and CCN5. Calcium fluorescent imaging demonstrated an increase in intracellular Ca2+ concentration upon stimulation with glutamate and KCl. Although neuronal features were induced similarly in basement membrane extract droplet culture, they diminished after culturing without SMs or in vivo transplantation into an injured spinal cord. In conclusion, SMs temporarily induce neuronal features in ACDFs. However, the analysis of bottlenecks in the neuronal induction is crucial for optimizing the process.


Subject(s)
Neurons , Spinal Cord , Animals , Dogs , Adult , Humans , Neurons/metabolism , Tubulin/metabolism , Fibroblasts/metabolism , Cell Differentiation/physiology , Cells, Cultured
3.
J Bone Miner Metab ; 40(4): 561-570, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35428898

ABSTRACT

BACKGROUND: Profilin-1 (Pfn1), an evolutionarily conserved actin-binding protein, is an important regulator of the cytoskeleton. We previously reported the osteoclast-specific Pfn1-conditional knockout (cKO) mice had postnatal osteolytic phenotype with craniofacial and long-bone deformities associated with increased migration of cultured osteoclasts. We hypothesized the increased cellular processes structured with branched actin filaments may underlies the mechanism of increased bone resorption in these mutant mice. MATERIALS AND METHODS: The morphological structure and cell migration of the cultured osteoclasts were analyzed using fluorescent microscopy and time-lapse image capturing. Fractional migration distances, as well as the index of protrusive structures (%-PB) that evaluates relative border length of the protrusion were compared between the cells from control and Pfn1-cKO mice. RESULTS: Time-lapse image analysis showed that %-PB was significantly larger in Pfn1-cKO osteoclasts. In addition, the fractional migration distance was positively correlated with the index. When the branched actin filament organization was suppressed by chemical inhibitors, the osteoclast migration was declined. Importantly, the suppression was more extensive in Pfn1-cKO than in control osteoclasts. CONCLUSION: Our results indicated the causative involvement of the increased branched actin filament formation at least in part for their excessive migration. Our findings provide a mechanistic rationale for testing novel therapeutic approaches targeting branched actin filaments in osteolytic disorders.


Subject(s)
Osteoclasts , Profilins , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Bone and Bones/metabolism , Cell Movement , Mice , Osteoclasts/metabolism , Profilins/genetics , Profilins/metabolism
5.
J Cell Physiol ; 233(1): 259-268, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28233307

ABSTRACT

Osteocytes are the most abundant cells in bone and regulate bone metabolism in coordination with osteoblasts and osteoclasts. However, the molecules that control osteocytes are still incompletely understood. Profilin1 is an actin-binding protein that is involved in actin polymerization. Osteocytes possess characteristic dendritic process formed based on actin cytoskeleton. Here, we examined the expression of profilin1 and its function in osteocytes. Profilin1 mRNA was expressed in osteocytic MLO-Y4 cells and its levels were gradually increased along with the time in culture. With regard to functional aspect, knockdown of profilin1 by siRNA enhanced BMP-induced increase in alkaline phosphatase expression levels in MLO-Y4 cells. Profilin1 knockdown suppressed the levels of dendritic processes and migration of MLO-Y4 cells. Since aging causes an increase in ROS in the body, we further examined the effects of hydrogen peroxide on the expression of profilin1. Hydrogen peroxide treatment increased the levels of profilin1 mRNA in MLO-Y4 cells in contrast to the decline in alkaline phosphatase. Profilin1 was expressed not only in MLO-Y4cells but also in the primary cultures of osteocytes. Importantly, profilin1 mRNA levels in primary cultures of osteocytes were higher than those in primary cultures of osteoblasts. To examine in vivo role of profilin1 in osteocytes, profilin1 was conditionally knocked out by using DMP1-cre and profilin1 floxed mice. This conditional deletion of profilin1 specifically in osteocytes resulted in reduction in the levels of bone volume and bone mineral density. These data indicate that profilin1 is expressed in osteocytes and regulates cell shape, migration and bone mass.


Subject(s)
Cell Movement , Cell Shape , Femur/metabolism , Osteocytes/metabolism , Profilins/metabolism , Alkaline Phosphatase/metabolism , Animals , Bone Density , Bone Remodeling , Cell Line , Cell Movement/drug effects , Cell Shape/drug effects , Femur/diagnostic imaging , Femur/drug effects , Gene Expression Regulation , Genotype , Hydrogen Peroxide/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Osteocytes/drug effects , Phenotype , Primary Cell Culture , Profilins/deficiency , Profilins/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Time Factors , Transfection , X-Ray Microtomography
7.
Proc Natl Acad Sci U S A ; 112(50): 15432-7, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26621720

ABSTRACT

Migration of the cells in osteoblastic lineage, including preosteoblasts and osteoblasts, has been postulated to influence bone formation. However, the molecular bases that link preosteoblastic/osteoblastic cell migration and bone formation are incompletely understood. Nck (noncatalytic region of tyrosine kinase; collectively referred to Nck1 and Nck2) is a member of the signaling adaptors that regulate cell migration and cytoskeletal structures, but its function in cells in the osteoblastic lineage is not known. Therefore, we examined the role of Nck in migration of these cells. Nck is expressed in preosteoblasts/osteoblasts, and its knockdown suppresses migration as well as cell spreading and attachment to substrates. In contrast, Nck1 overexpression enhances spreading and increases migration and attachment. As for signaling, Nck double knockdown suppresses migration toward IGF1 (insulin-like growth factor 1). In these cells, Nck1 binds to IRS-1 (insulin receptor substrate 1) based on immunoprecipitation experiments using anti-Nck and anti-IRS-1 antibodies. In vivo, Nck knockdown suppresses enlargement of the pellet of DiI-labeled preosteoblasts/osteoblasts placed in the calvarial defects. Genetic experiments indicate that conditional double deletion of both Nck1 and Nck2 specifically in osteoblasts causes osteopenia. In these mice, Nck double deficiency suppresses the levels of bone-formation parameters such as bone formation rate in vivo. Interestingly, bone-resorption parameters are not affected. Finally, Nck deficiency suppresses repair of bone injury after bone marrow ablation. These results reveal that Nck regulates preosteoblastic/osteoblastic migration and bone mass.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bone and Bones/cytology , Cell Movement , Oncogene Proteins/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Animals , Bone Resorption/metabolism , Bone Resorption/pathology , Bone and Bones/diagnostic imaging , Bone and Bones/drug effects , Cell Movement/drug effects , Cell Shape/drug effects , Cells, Cultured , Gene Knockdown Techniques , Insulin Receptor Substrate Proteins/metabolism , Insulin-Like Growth Factor I/pharmacology , Mice, Knockout , Oncogene Proteins/deficiency , Organ Size , Osteoblasts/drug effects , Osteogenesis/drug effects , Protein Binding/drug effects , Radiography , Skull/drug effects , Skull/metabolism , Wound Healing/drug effects
8.
J Cell Physiol ; 232(7): 1761-1766, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27861872

ABSTRACT

LGR4 is expressed in bone and has been shown to be involved in bone metabolism. Oxidative stress is one of the key issues in pathophysiology of osteoporosis. However, the link between Lgr4 and oxidative stress has not been known. Therefore, effects of hydrogen peroxide on Lgr4 expression in osteoblasts were examined. Hydrogen peroxide treatment suppressed the levels of Lgr4 mRNA expression in an osteoblastic cell line, MC3T3-E1. The suppressive effects were not obvious at 0.1 mM, while 1 mM hydrogen peroxide suppressed Lgr4 expression by more than 50%. Hydrogen peroxide treatment suppressed Lgr4 expression within 12 h and this suppression lasted at least up to 48 h. Hydrogen peroxide suppression of Lgr4 expression was still observed in the presence of a transcription inhibitor but was no longer observed in the presence of a protein synthesis inhibitor. Although Lgr4 expression in osteoblasts is enhanced by BMP2 treatment as reported before, hydrogen peroxide treatment suppressed Lgr4 even in the presence of BMP2. Finally, hydrogen peroxide suppressed Lgr4 expression in primary cultures of osteoblasts similarly to MC3T3-E1 cells. These date indicate that hydrogen peroxide suppresses Lgr4 expression in osteoblastic cells. J. Cell. Physiol. 232: 1761-1766, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Hydrogen Peroxide/toxicity , Osteoblasts/metabolism , Receptors, G-Protein-Coupled/metabolism , Alkaline Phosphatase/metabolism , Animals , Cell Line , Cytokines/pharmacology , Down-Regulation/drug effects , Mice , Osteoblasts/drug effects , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Receptors, G-Protein-Coupled/genetics , Time Factors , Transcription, Genetic/drug effects
9.
J Cell Biochem ; 118(7): 1670-1677, 2017 07.
Article in English | MEDLINE | ID: mdl-27918072

ABSTRACT

Osteoporosis is one of the most prevalent ageing-associated diseases that are soaring in the modern world. Although various aspects of the disease have been investigated to understand the bases of osteoporosis, the pathophysiological mechanisms underlying bone loss is still incompletely understood. Poldip2 is a molecule that has been shown to be involved in cell migration of vascular cells and angiogenesis. However, expression of Poldip2 and its regulation in bone cells were not known. Therefore, we examined the Poldip2 mRNA expression and the effects of bone regulators on the Poldip2 expression in osteoblasts. We found that Poldip2 mRNA is expressed in osteoblastic MC3T3-E1 cells. As FGF controls osteoblasts and angiogenesis, FGF regulation was investigated in these cells. FGF suppressed the expression of Poldip2 in MC3T3-E1 cells in a time dependent manner. Protein synthesis inhibitor but not transcription inhibitor reduced the FGF effects on Poldip2 gene expression in MC3T3-E1 cells. As for bone-related hormones, dexamethasone was found to enhance the expression of Poldip2 in osteoblastic MC3T3-E1 cells whereas FGF still suppressed such dexamethasone effects. With respect to function, knockdown of Poldip2 by siRNA suppressed the migration of MC3T3-E1 cells. Poldip2 was also expressed in the primary cultures of osteoblast-enriched cells and FGF also suppressed its expression. Finally, Poldip2 was expressed in femoral bone in vivo and its levels were increased in aged mice compared to young adult mice. These data indicate that Poldip2 is expressed in osteoblastic cells and is one of the targets of FGF. J. Cell. Biochem. 118: 1670-1677, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Fibroblast Growth Factors/pharmacology , Mitochondrial Proteins/metabolism , Nuclear Proteins/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Animals , Cell Line , Cell Movement/drug effects , Cell Movement/genetics , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , Nuclear Proteins/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction
10.
J Cell Physiol ; 231(2): 496-504, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26192605

ABSTRACT

Osteoporosis affects over 20 million patients in the United States. Among those, disuse osteoporosis is serious as it is induced by bed-ridden conditions in patients suffering from aging-associated diseases including cardiovascular, neurological, and malignant neoplastic diseases. Although the phenomenon that loss of mechanical stress such as bed-ridden condition reduces bone mass is clear, molecular bases for the disuse osteoporosis are still incompletely understood. In disuse osteoporosis model, bone loss is interfered by inhibitors of sympathetic tone and adrenergic receptors that suppress bone formation. However, how beta adrenergic stimulation affects osteoblastic migration and associated proliferation is not known. Here we introduced a live imaging system, fluorescent ubiquitination-based cell cycle indicator (FUCCI), in osteoblast biology and examined isoproterenol regulation of cell cycle transition and cell migration in osteoblasts. Isoproterenol treatment suppresses the levels of first entry peak of quiescent osteoblastic cells into cell cycle phase by shifting from G1 /G0 to S/G2 /M and also suppresses the levels of second major peak population that enters into S/G2 /M. The isoproterenol regulation of osteoblastic cell cycle transition is associated with isoproterenol suppression on the velocity of migration. This isoproterenol regulation of migration velocity is cell cycle phase specific as it suppresses migration velocity of osteoblasts in G1 phase but not in G1 /S nor in G2 /M phase. Finally, these observations on isoproterenol regulation of osteoblastic migration and cell cycle transition are opposite to the PTH actions in osteoblasts. In summary, we discovered that sympathetic tone regulates osteoblastic migration in association with cell cycle transition by using FUCCI system.


Subject(s)
Osteoblasts/cytology , Osteoblasts/metabolism , Receptors, Adrenergic, beta/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Cell Cycle Checkpoints , Cell Movement/drug effects , Cells, Cultured , Isoproterenol/pharmacology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Osteoblasts/drug effects , Parathyroid Hormone/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Single-Cell Analysis
11.
J Cell Physiol ; 231(4): 887-95, 2016 04.
Article in English | MEDLINE | ID: mdl-26332449

ABSTRACT

Osteoporosis is one of the most prevalent diseases and the number of patients suffering from this disease is soaring due to the increase in the aged population in the world. The severity of bone loss in osteoporosis is based on the levels of impairment in the balance between bone formation and bone resorption, two arms of the bone metabolism, and bone remodeling. However, determination of bone formation levels is under many layers of control that are as yet fully defined. Bone morphogenetic protein (BMP) plays a key role in regulation of bone formation while its downstream targets are still incompletely understood. Lgr4 gene encodes an orphan receptor and has been identified as a genetic determinant for bone mass in osteoporotic patients. Here, we examine the effects of BMP on the expression of Lgr4 in osteoblastic cells. Lgr4 gene is expressed in an osteoblastic cell line, MC3T3E1 in a time dependent manner during the culture. BMP treatment enhances Lgr4 mRNA expression at least in part via transcriptional event. When Lgr4 mRNA is knocked down, the levels of BMP-induced increase in alkaline phosphatase (Alp) activity and Alp mRNA are suppressed. BMP enhancement of Lgr4 gene expression is suppressed by FGF and reversed by dexamethasone. BMP also enhances Lgr4 expression in primary cultures of calvarial osteoblasts. These data indicate that Lgr4 gene is regulated by BMP and is required for BMP effects on osteoblastic differentiation. J. Cell. Physiol. 231: 887-895, 2016. © 2015 Wiley Periodicals, Inc.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Gene Expression Regulation/drug effects , Osteoblasts/metabolism , Receptors, G-Protein-Coupled/genetics , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Bone and Bones/drug effects , Bone and Bones/metabolism , Cell Line , Gene Knockdown Techniques , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/enzymology , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/metabolism , Time Factors , Transcription, Genetic/drug effects
12.
J Cell Physiol ; 231(5): 1163-70, 2016 May.
Article in English | MEDLINE | ID: mdl-26460818

ABSTRACT

Unloading induces bone loss and causes disuse osteoporosis. However, the mechanism underlying disuse osteoporosis is still incompletely understood. Here, we examined the effects of cathepsin K (CatK) deficiency on disuse osteoporosis induced by using sciatic neurectomy (Nx) model. After 4 weeks of surgery, CatK KO and WT mice were sacrificed and subjected to analyses. For cancellous bone rich region, Nx reduced the bone mineral density (BMD) compared to the BMD in the sham operated side in wild type mice. In contrast, CatK deficiency suppressed such Nx-induced reduction of BMD in cancellous bone. Nx also reduced BMD in the mid shaft cortical bone compared to the BMD in the corresponding region on the sham operated side in wild type mice. In contrast, CatK deficiency suppressed such Nx-induced reduction of BMD in the mid shaft cortical bone. Bone volume (BV/TV) was reduced by Nx in WT mice. In contrast, Cat-K deficiency suppressed such reduction in bone volume. Interestingly, CatK deficiency suppressed osteoclast number and osteoclast surface in the Nx side compared to sham side. When bone marrow cells obtained from Nx side femur of CatK-KO mice were cultured, the levels of the calcified area in culture were increased. Further examination of gene expression indicated that Nx suppressed the expression of genes encoding osteoblast-phenotype-related molecules such as Runx2 and alkaline phosphatase in WT mice. In contrast, CatK deficiency suppressed such reduction. These data indicate that CatK is involved in the disuse-induced bone mass reduction.


Subject(s)
Bone Resorption/enzymology , Bone Resorption/etiology , Cathepsin K/deficiency , Muscular Disorders, Atrophic/complications , Muscular Disorders, Atrophic/enzymology , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Bone Density , Bone Marrow Cells/metabolism , Bone Resorption/diagnostic imaging , Bone Resorption/physiopathology , Bone and Bones/diagnostic imaging , Bone and Bones/enzymology , Bone and Bones/pathology , Calcification, Physiologic/genetics , Cathepsin K/metabolism , Cells, Cultured , Imaging, Three-Dimensional , Mice, Inbred C57BL , Muscular Disorders, Atrophic/diagnostic imaging , Muscular Disorders, Atrophic/pathology , Organ Size , Osteoclasts/pathology , Osteogenesis/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , X-Ray Microtomography
13.
BMC Genomics ; 17(1): 979, 2016 11 28.
Article in English | MEDLINE | ID: mdl-27894274

ABSTRACT

BACKGROUND: Although CRISPR/Cas enables one-step gene cassette knock-in, assembling targeting vectors containing long homology arms is a laborious process for high-throughput knock-in. We recently developed the CRISPR/Cas-based precise integration into the target chromosome (PITCh) system for a gene cassette knock-in without long homology arms mediated by microhomology-mediated end-joining. RESULTS: Here, we identified exonuclease 1 (Exo1) as an enhancer for PITCh in human cells. By combining the Exo1 and PITCh-directed donor vectors, we achieved convenient one-step knock-in of gene cassettes and floxed allele both in human cells and mouse zygotes. CONCLUSIONS: Our results provide a technical platform for high-throughput knock-in.


Subject(s)
Gene Knock-In Techniques , Homologous Recombination , Zygote , Animals , Base Sequence , CRISPR-Cas Systems , Cell Line , Chromosomes , Clustered Regularly Interspaced Short Palindromic Repeats , Exodeoxyribonucleases/metabolism , Gene Targeting , Genetic Loci , Humans , Mice , Transcription Activator-Like Effector Nucleases
14.
J Cell Biochem ; 117(4): 970-7, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26378628

ABSTRACT

CIZ/NMP4 (Cas interacting zinc finger protein, Nmp4, Zfp384) is a transcription factor that is known to regulate matrix related-proteins. To explore the possible pathophysiological role of CIZ/NMP4 in arthritis, we examined CIZ/NMP4 expression in articular cartilage in arthritis model. CIZ/NMP4 was expressed in the articular chondrocytes of mice at low levels while its expression was enhanced when arthritis was induced. Arthritis induction increased clinical score in wild type mice. In contrast, CIZ/NMP4 deficiency suppressed such rise in the levels of arthritis score and swelling of soft tissue. CIZ/NMP4 deficiency also reduced invasion of inflammatory cells in joint tissue. Quantitative PCR analyses of mRNA from joints revealed that arthritis-induced increase in expressions of IL-1ß was suppressed by CIZ/NMP4 deficiency. CIZ/NMP4 bound to IL-1ß promoter and activated its transcription. The increase in CIZ/NMP4 in arthritis was also associated with enhancement in bone resorption and cartilage matrix degradation. In fact, RANKL, a signaling molecule prerequisite for osteoclastogenesis and, MMP-3, a clinical marker for arthritis were increased in joints upon arthritis induction. In contrast, CIZ/NMP4 deficiency suppressed the arthritis-induced increase in bone resorption, expression of RANKL and MMP-3 mRNA. Thus, CIZ/NMP4 plays a role in the development of arthritis at least in part through regulation of key molecules related to the arthritis.


Subject(s)
Arthritis, Experimental/genetics , Cartilage, Articular/immunology , Matrix Metalloproteinase 3/immunology , Nuclear Matrix-Associated Proteins/immunology , RANK Ligand/immunology , Transcription Factors/immunology , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Autoantibodies/biosynthesis , Bone Resorption , Cartilage, Articular/pathology , Chondrocytes/immunology , Chondrocytes/pathology , Female , Gene Expression Regulation , Glucose-6-Phosphate Isomerase/antagonists & inhibitors , Glucose-6-Phosphate Isomerase/genetics , Glucose-6-Phosphate Isomerase/immunology , Immune Sera/administration & dosage , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Joints/immunology , Joints/pathology , Male , Matrix Metalloproteinase 3/genetics , Mice , Mice, Knockout , Nuclear Matrix-Associated Proteins/deficiency , Nuclear Matrix-Associated Proteins/genetics , Promoter Regions, Genetic , RANK Ligand/genetics , Severity of Illness Index , Signal Transduction , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic
15.
J Cell Biochem ; 117(3): 621-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26271366

ABSTRACT

Profilin 1 (Pfn1) regulates cytoskeletal reorganization and migration, but its role in osteoblasts is not known. BMP (bone morphogenetic protein) is a multifunctional cytokine involved in osteoblastic differentiation and promotes bone regeneration and repair. Although several molecules are known to modulate BMP signaling, mechanisms that determine the levels of BMP action in osteoblastic function are still incompletely understood. We therefore examine the expression of Pfn1 in osteoblasts and its role in BMP-induced differentiation in osteoblasts. In osteoblastic MC3T3-E1(MC) cells, Pfn1 mRNA is expressed constitutively and its expression levels are declined during the culture in a time dependent manner in contrast to the increase in alkaline phosphatase activity revealing that Pfn1 expression is down regulated along with differentiation. To test the effects of osteoblastic differentiation on Pfn1expression further, MC cells are treated with BMP. BMP treatment suppresses the levels of Pfn1 mRNA. This suppressive effect of BMP is time dependent and further down regulation of Pfn1 mRNA levels is observed when the BMP treatment is continued for a longer period of time. Pfn1mRNA knock down (KD) by siRNAs enhances BMP-induced increase in alkaline phosphatase (Alp) activity in MC cells. To analyze the regulatory mechanism, Alp mRNA levels are examined and Pfn1 KD enhances the BMP-induced increase in the levels of Alp mRNA expression. Furthermore, Pfn1 KD enhances BMP-induced transcriptional expression of luciferase reporter activity via BMP response element in osteoblasts. These data indicate that Pfn1 is a novel target of BMP and suppresses BMP-induced differentiation of osteoblasts at least in part via transcriptional event.


Subject(s)
Osteoblasts/metabolism , Profilins/metabolism , 3T3 Cells , Alkaline Phosphatase/metabolism , Animals , Bone Morphogenetic Protein 2/physiology , Enzyme Induction , Gene Silencing , Mice , Profilins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Response Elements , Transcription, Genetic
16.
Hum Mol Genet ; 23(9): 2339-52, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24334604

ABSTRACT

Collagen VI-related myopathies are disorders of connective tissue presenting with an overlap phenotype combining clinical involvement from the muscle and from the connective tissue. Not all patients displaying related overlap phenotypes between muscle and connective tissue have mutations in collagen VI. Here, we report a homozygous recessive loss of function mutation and a de novo dominant mutation in collagen XII (COL12A1) as underlying a novel overlap syndrome involving muscle and connective tissue. Two siblings homozygous for a loss of function mutation showed widespread joint hyperlaxity combined with weakness precluding independent ambulation, while the patient with the de novo missense mutation was more mildly affected, showing improvement including the acquisition of walking. A mouse model with inactivation of the Col12a1 gene showed decreased grip strength, a delay in fiber-type transition and a deficiency in passive force generation while the muscle seems more resistant to eccentric contraction induced force drop, indicating a role for a matrix-based passive force-transducing elastic element in the generation of the weakness. This new muscle connective tissue overlap syndrome expands on the emerging importance of the muscle extracellular matrix in the pathogenesis of muscle disease.


Subject(s)
Collagen Type XII/genetics , Muscular Diseases/genetics , Mutation/genetics , Animals , Child, Preschool , Collagen Type VI/genetics , Collagen Type VI/metabolism , Collagen Type XII/metabolism , Disease Models, Animal , Humans , Infant , Male , Mice , Muscle, Skeletal/pathology , Muscular Diseases/metabolism , Muscular Diseases/pathology
17.
Cell Tissue Res ; 364(3): 623-635, 2016 06.
Article in English | MEDLINE | ID: mdl-26753503

ABSTRACT

Bone formation is precisely regulated by cell-cell communication in osteoblasts. We have previously demonstrated that genetic deletion of Col6a1 or Col12a1 impairs osteoblast connections and/or communication in mice, resulting in bone mass reduction and bone fragility. Mutations of the genes encoding collagen VI cause Ullrich congenital muscular dystrophy (UCMD) and Bethlem myopathy (BM), which have overlapping phenotypes involving connective tissue and muscle. Recent studies have identified COL12A1 gene mutations in patients with UCMD- and BM-like disorders harboring no COL6 mutations, indicating the shared functions of these collagens in connective tissue homeostasis. The purpose of this investigation has been to test the hypothesis that collagens VI and XII have coordinate regulatory role(s) during bone formation. We analyzed the localization of collagens VI and XII relative to primary osteoblasts during osteogenesis. Immunofluorescence analysis demonstrated that collagens VI and XII colocalized in matrix bridges between adjacent cells during periods when osteoblasts were establishing cell-cell connections. Quantification of cells harboring collagen bridges demonstrated that matrix bridges were composed of collagens VI and XII but not collagen I. Interestingly, matrix bridge formation was impaired in osteoblasts deficient in either Col6a1 or Col12a1, suggesting that both collagens were indispensable for matrix bridge formation. These data demonstrate, for the first time, a functional relationship between collagens VI and XII during osteogenesis and indicate that a complex containing collagens VI and XII is essential for the formation of a communicating cellular network during bone formation.


Subject(s)
Cell Communication , Collagen Type VI/metabolism , Collagen Type XII/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis , Animals , Cells, Cultured , Collagen Type I/metabolism , Culture Media/pharmacology , Extracellular Matrix/metabolism , Mice , Protein Binding
18.
Calcif Tissue Int ; 99(2): 199-208, 2016 08.
Article in English | MEDLINE | ID: mdl-27086348

ABSTRACT

Heterotopic ossification (HO) in various tissues evokes clinical problems. Inflammatory responses of the stromal progenitor cells may be involved in its etiology. Previous report indicated that pro-inflammatory cytokines including IL-1ß enhanced the in vitro calcification of human mesenchymal stem cells (MSCs), by suppressing the expression of ectonucleotide pyrophosphatase/phosphodiesterase-1 gene (ENPP1). However, possible contribution of other related factors had not been investigated. Here, we investigated the expression of regulators of extracellular pyrophosphate and nucleosides including Enpp1, Nt5e, Ank, Enptds, and Ent1, examining various connective tissue stromal progenitor cells, including bone marrow stromal cells and synovium derived cells from mouse, or bone marrow MSCs from human. Consistent with previous studies, we observed characteristic suppression of the osteoblastic marker genes by IL-1ß during the osteogenic culture for 20 days. In addition, we observed a reduced expression of the important transporter genes, Ank and Ent1, whereas the alteration in Enpp1 and Nt5e levels was not always consistent among the cell types. Our results suggest that IL-1ß suppresses not only the osteoblastic but also the negative regulators of soft-tissue calcification, including Ank and Ent1 in stromal progenitor cells, which may contribute to the mechanisms of HO in various disorders.


Subject(s)
Cell Differentiation/drug effects , Equilibrative Nucleoside Transporter 1/metabolism , Interleukin-1beta/pharmacology , Mesenchymal Stem Cells/drug effects , Osteogenesis/drug effects , Phosphate Transport Proteins/metabolism , Stromal Cells/drug effects , Animals , Calcification, Physiologic/drug effects , Calcification, Physiologic/physiology , Calcinosis/metabolism , Cell Differentiation/physiology , Cells, Cultured , Humans , Interleukin-1beta/metabolism , Mesenchymal Stem Cells/cytology , Mice , Osteogenesis/physiology , Stem Cells/drug effects , Stem Cells/metabolism , Stromal Cells/cytology
19.
J Cell Physiol ; 230(11): 2788-95, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25828538

ABSTRACT

Ift88 is an intraflagella transport protein, critical for the cilium, and has been shown to be required for the maintenance of chondrocytes and cartilage. However, how Ift88 is controlled by cytokines that play a role in osteoarthritis is not well understood. Therefore, we examined the effects of TGF-ß on the expression of Ift88. We used ATDC5 cells as chondrocytes and analyzed the effects of TGF-ß on gene expression. TGF-ß treatment suppresses the levels of Ift88 mRNA in a dose-dependent manner starting from as low as 0.5 ng/mL and reaching the nadir at around 2 ng/mL. TGF-ß treatment also suppresses the protein levels of Ift88. TGF-ß suppression of Ift88 is still observed when the cells are cultured in the presence of a transcriptional inhibitor while the TGF-ß suppression is weakened in the presence of a protein synthesis inhibitor, cycloheximide. TGF-ß treatment suppresses the levels of Ift88 mRNA stability suggesting the presence of posttranscriptional regulation. TGF-ß treatment reduces the number of cilia positive cells and suppresses average length of cilia. Knockdown of Ift88 by siRNA enhances TGF-ß-induced increase in type II collagen mRNA expression in ATDC5 cells revealing the suppressive role of Ift88 on TGF-ß-induced regulation of extracellular matrix protein expression. TGF-ß also suppresses Ift88 mRNA expression in primary culture of rib chondrocytes. These data indicate that TGF-ß regulates Ift88 gene expression at least in part via posttrascriptional manner.


Subject(s)
Cilia/metabolism , Osteoarthritis/genetics , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/biosynthesis , Animals , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Cilia/pathology , Gene Expression Regulation/drug effects , Mice , Osteoarthritis/metabolism , Osteoarthritis/pathology , Phosphorylation , Signal Transduction , Transforming Growth Factor beta/administration & dosage , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
20.
J Cell Biochem ; 116(6): 1144-52, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25536656

ABSTRACT

ß adrenergic stimulation suppresses bone formation in vivo while its actions in osteoblastic differentiation are still incompletely understood. We therefore examined the effects of ß2 adrenergic stimulation on osteoblast-like MC3T3-E1 cells focusing on BMP-induced alkaline phosphatase expression. Morphologically, isoproterenol treatment suppresses BMP-induced increase in the numbers of alkaline phosphatase-positive small foci in the cultures of MC3T3-E1 cells. Biochemically, isoproterenol treatment suppresses BMP-induced enzymatic activity of alkaline phosphatase in a dose-dependent manner. Isoproterenol suppression of alkaline phosphatase activity is observed even when the cells are treated with high concentrations of BMP. With respect to cell density, isoproterenol treatment tends to suppress BMP-induced increase in alkaline phosphatase expression more in osteoblasts cultured at higher cell density. In terms of treatment protocol, continuous isoproterenol treatment is compared to cyclic treatment. Continuous isoproterenol treatment is more suppressive against BMP-induced increase in alkaline phosphatase expression than cyclic regimen. At molecular level, isoproterenol treatment suppresses BMP-induced enhancement of alkaline phosphatase mRNA expression. Regarding the mode of isoproterenol action, isoproterenol suppresses BMP-induced BRE-luciferase activity. These data indicate that isoproterenol regulates BMP-induced alkaline phosphatase expression in osteoblast-like MC3T3E1 cells.


Subject(s)
Alkaline Phosphatase/metabolism , Bone Morphogenetic Proteins/pharmacology , Osteoblasts/metabolism , Receptors, Adrenergic/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Mice , Signal Transduction/drug effects , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL