Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(17)2023 Sep 04.
Article in English | MEDLINE | ID: mdl-37686441

ABSTRACT

The vast majority of transcribed RNAs are noncoding RNAs. Among noncoding RNAs, long noncoding RNAs (lncRNAs), which contain hundreds to thousands of bases, have received attention in many fields. The vast majority of the constituent cells in bone tissue are osteocytes, but their regulatory mechanisms are incompletely understood. Considering the wide range of potential contributions of lncRNAs to physiological processes and pathological conditions, we hypothesized that lncRNAs in osteocytes, which have not been reported, could be involved in bone metabolism. Here, we first isolated osteocytes from femurs of mice with osteocyte-specific GFP expression. Then, through RNA-sequencing, we identified osteocyte-specific lncRNAs and focused on a novel lncRNA, 9530026P05Rik (lncRNA953Rik), which strongly suppressed osteogenic differentiation. In the IDG-SW3 osteocyte line with lncRNA953Rik overexpression, the expression of Osterix and its downstream genes was reduced. RNA pull-down and subsequent LC-MS/MS analysis revealed that lncRNA953Rik bound the nuclear protein CCAR2. We demonstrated that CCAR2 promoted Wnt/ß-catenin signaling and that lncRNA953Rik inhibited this pathway. lncRNA953Rik sequestered CCAR2 from HDAC1, leading to deacetylation of H3K27 in the Osterix promoter and consequent transcriptional downregulation of Osterix. This research is the first to clarify the role of a lncRNA in osteocytes. Our findings can pave the way for novel therapeutic options targeting lncRNAs in osteocytes to treat bone metabolic diseases such as osteoporosis.


Subject(s)
Osteocytes , Osteogenesis , RNA, Long Noncoding , Animals , Mice , Chromatography, Liquid , Osteogenesis/genetics , RNA, Long Noncoding/genetics , Tandem Mass Spectrometry , Wnt Signaling Pathway
2.
Sci Rep ; 13(1): 4674, 2023 03 22.
Article in English | MEDLINE | ID: mdl-36949102

ABSTRACT

Bone is one of the largest organ systems in humans and is considered to regulate whole-body homeostasis in cooperation with other organs. We have previously reported that a sympathetic or sensory nervous system inside bone regulates bone homeostasis. However, the detailed regulatory mechanism, including the distribution of nerves inside bone, remains unknown. Although a two-dimensional histological analysis has been widely used to evaluate the structure of nerves or blood vessels, the actual structure is more complex, suggesting that it should be evaluated three-dimensionally. Here, we established a novel bone tissue clearing technique (Osteo-DISCO) for murine bones which enabled us to visualize the detailed distribution of nerves or blood vessels inside bone. Interestingly, we found that there is a specific nerve entry site in each long bone and that surgical ablation of the specific nerve fibers entering bone tissue led to decreased bone formation and impaired bone regeneration. Furthermore, we revealed that the administration of calcitonin gene-related peptide (CGRP), which is primarily released from sensory nerves, suppressed the bone loss caused by surgical nerve ablation. An in vitro study also indicated that CGRP directly promotes osteoblast activity, suggesting that sensory nerves inside bone can regulate osteogenesis via the secretion of CGRP.


Subject(s)
Calcitonin Gene-Related Peptide , Imaging, Three-Dimensional , Mice , Humans , Animals , Bone and Bones/diagnostic imaging , Bone Remodeling , Neural Networks, Computer
3.
Int J Mol Sci ; 20(5)2019 Mar 02.
Article in English | MEDLINE | ID: mdl-30832329

ABSTRACT

Pericytes are mesenchymal cells that surround the endothelial cells of small vessels in various organs. These cells express several markers, such as NG2, CD146, and PDGFRß, and play an important role in the stabilization and maturation of blood vessels. It was also recently revealed that like mesenchymal stem cells (MSCs), pericytes possess multilineage differentiation capacity, especially myogenic, adipogenic, and fibrogenic differentiation capacities. Although some previous studies have reported that pericytes also have osteogenic potential, the osteogenesis of pericytes can still be further elucidated. In the present study, we established novel methods for isolating and culturing primary murine pericytes. An immortalized pericyte line was also established. Multilineage induction of the pericyte line induced osteogenesis, adipogenesis, and chondrogenesis of the cells in vitro. In addition, pericytes that were injected into the fracture site of a bone fracture mouse model contributed to callus formation. Furthermore, in vivo pericyte-lineage-tracing studies demonstrated that endogenous pericytes also differentiate into osteoblasts and osteocytes and contribute to bone fracture healing as a cellular source of osteogenic cells. Pericytes can be a promising therapeutic candidate for treating bone fractures with a delayed union or nonunion as well as bone diseases causing bone defects.


Subject(s)
Chondrogenesis , Fracture Healing , Osteogenesis , Pericytes/cytology , Primary Cell Culture/methods , Animals , Cell Differentiation , Cell Line , Cells, Cultured , Chondrocytes/cytology , Mesenchymal Stem Cell Transplantation/methods , Mice , Mice, Inbred BALB C , Osteoblasts/cytology , Pericytes/transplantation
4.
Proc Natl Acad Sci U S A ; 115(9): 2204-2209, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29440427

ABSTRACT

Bone metastatic lesions are classified as osteoblastic or osteolytic lesions. Prostate and breast cancer patients frequently exhibit osteoblastic-type and osteolytic-type bone metastasis, respectively. In metastatic lesions, tumor cells interact with many different cell types, including osteoblasts, osteoclasts, and mesenchymal stem cells, resulting in an osteoblastic or osteolytic phenotype. However, the mechanisms responsible for the modification of bone remodeling have not been fully elucidated. MicroRNAs (miRNAs) are transferred between cells via exosomes and serve as intercellular communication tools, and numerous studies have demonstrated that cancer-secreted miRNAs are capable of modifying the tumor microenvironment. Thus, cancer-secreted miRNAs can induce an osteoblastic or osteolytic phenotype in the bone metastatic microenvironment. In this study, we performed a comprehensive expression analysis of exosomal miRNAs secreted by several human cancer cell lines and identified eight types of human miRNAs that were highly expressed in exosomes from osteoblastic phenotype-inducing prostate cancer cell lines. One of these miRNAs, hsa-miR-940, significantly promoted the osteogenic differentiation of human mesenchymal stem cells in vitro by targeting ARHGAP1 and FAM134A Interestingly, although MDA-MB-231 breast cancer cells are commonly known as an osteolytic phenotype-inducing cancer cell line, the implantation of miR-940-overexpressing MDA-MB-231 cells induced extensive osteoblastic lesions in the resulting tumors by facilitating the osteogenic differentiation of host mesenchymal cells. Our results suggest that the phenotypes of bone metastases can be induced by miRNAs secreted by cancer cells in the bone microenvironment.


Subject(s)
Bone Neoplasms/metabolism , Breast Neoplasms/pathology , GTPase-Activating Proteins/metabolism , Membrane Proteins/metabolism , MicroRNAs/metabolism , Prostatic Neoplasms/metabolism , Adenocarcinoma/metabolism , Animals , Bone Neoplasms/secondary , Bone Substitutes , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , GTPase-Activating Proteins/genetics , Humans , Male , Membrane Proteins/genetics , Mesenchymal Stem Cells , Mice , MicroRNAs/genetics , Neoplasms, Experimental/metabolism
5.
J Infect Dis ; 215(12): 1893-1897, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28525596

ABSTRACT

Long-term antiretroviral therapy is associated with increased fracture risk, but the mechanism remains elusive. We measured serum undercarboxylated osteocalcin and pentosidine (markers of poor bone quality) in human immunodeficiency virus-infected patients treated with protease inhibitors (PIs) or an integrase strand transfer inhibitor-containing regimen. The results demonstrated significantly higher undercarboxylated osteocalcin and pentosidine in PI-treated patients. Switching to integrase strand transfer inhibitor significant decreased these markers. We also showed impaired bone mechanical properties with higher undercarboxylated osteocalcin level in PI-treated mice and inhibited osteoblast differentiation in PI-treated osteogenic cells. The results confirmed the adverse effects of PIs on bone quality and osteoblast differentiation.


Subject(s)
Antiretroviral Therapy, Highly Active/adverse effects , Bone Density/drug effects , Protease Inhibitors/adverse effects , Animals , Arginine/analogs & derivatives , Arginine/blood , Biomarkers/blood , Female , HIV-1/drug effects , Humans , Integrase Inhibitors , Lysine/analogs & derivatives , Lysine/blood , Male , Mice , Osteocalcin/blood , Retrospective Studies , Reverse Transcriptase Inhibitors
6.
JCI Insight ; 1(5): e85061, 2016 04 21.
Article in English | MEDLINE | ID: mdl-27699258

ABSTRACT

Multiple myeloma (MM) is a relapsed and refractory disease, one that highlights the need for developing new molecular therapies for overcoming of drug resistance. Addition of panobinostat, a histone deacetylase (HDAC) inhibitor, to bortezomib and dexamethasone improved progression-free survival (PFS) in relapsed and refractory MM patients. Here, we demonstrate how calcineurin, when inhibited by immunosuppressive drugs like FK506, is involved in myeloma cell growth and targeted by panobinostat. mRNA expression of PPP3CA, a catalytic subunit of calcineurin, was high in advanced patients. Panobinostat degraded PPP3CA, a degradation that should have been induced by inhibition of the chaperone function of heat shock protein 90 (HSP90). Cotreatment with HDAC inhibitors and FK506 led to an enhanced antimyeloma effect with a greater PPP3CA reduction compared with HDAC inhibitors alone both in vitro and in vivo. In addition, this combination treatment efficiently blocked osteoclast formation, which results in osteolytic lesions. The poor response and short PFS duration observed in the bortezomib-containing therapies of patients with high PPP3CA suggested its relevance to bortezomib resistance. Moreover, bortezomib and HDAC inhibitors synergistically suppressed MM cell viability through PPP3CA inhibition. Our findings underscore the usefulness of calcineurin-targeted therapy in MM patients, including patients who are resistant to bortezomib.


Subject(s)
Calcineurin/metabolism , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Indoles/therapeutic use , Multiple Myeloma/drug therapy , Animals , Cell Line, Tumor , Female , Humans , Mice, Inbred NOD , Mice, SCID , Panobinostat , Tacrolimus/pharmacology , Xenograft Model Antitumor Assays
7.
J Bone Miner Res ; 31(7): 1344-55, 2016 07.
Article in English | MEDLINE | ID: mdl-26841172

ABSTRACT

The circadian clock controls many behavioral and physiological processes beyond daily rhythms. Circadian dysfunction increases the risk of cancer, obesity, and cardiovascular and metabolic diseases. Although clinical studies have shown that bone resorption is controlled by circadian rhythm, as indicated by diurnal variations in bone resorption, the molecular mechanism of circadian clock-dependent bone resorption remains unknown. To clarify the role of circadian rhythm in bone resorption, aryl hydrocarbon receptor nuclear translocator-like (Bmal1), a prototype circadian gene, was knocked out specifically in osteoclasts. Osteoclast-specific Bmal1-knockout mice showed a high bone mass phenotype due to reduced osteoclast differentiation. A cell-based assay revealed that BMAL1 upregulated nuclear factor of activated T cells, cytoplasmic, calcineurin-dependent 1 (Nfatc1) transcription through its binding to an E-box element located on the Nfatc1 promoter in cooperation with circadian locomotor output cycles kaput (CLOCK), a heterodimer partner of BMAL1. Moreover, steroid receptor coactivator (SRC) family members were shown to interact with and upregulate BMAL1:CLOCK transcriptional activity. Collectively, these data suggest that bone resorption is controlled by osteoclastic BMAL1 through interactions with the SRC family and binding to the Nfatc1 promoter. © 2016 American Society for Bone and Mineral Research.


Subject(s)
ARNTL Transcription Factors/metabolism , Bone Resorption/metabolism , Circadian Clocks/physiology , Osteoclasts/metabolism , ARNTL Transcription Factors/genetics , Animals , Bone Resorption/genetics , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Cell Differentiation/physiology , Mice , Mice, Knockout , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , src-Family Kinases/genetics , src-Family Kinases/metabolism
8.
FEBS Lett ; 589(21): 3302-8, 2015 Oct 24.
Article in English | MEDLINE | ID: mdl-26450370

ABSTRACT

Osteoblastic differentiation is regulated by various factors, including hormones and transcription factors. Runt-related transcription factor 2 (Runx2) is an essential player in osteoblastogenesis and transactivates its molecular target by creating a protein complex with its hetero-dimeric partner core binding factor beta (Cbfb). However, the molecular regulation of Cbfb expression remains unknown. Here, we identified miR-145 as a crucial regulator of Cbfb expression. The expression of miR-145 increased during osteoblastogenesis, indicating that miR-145 works as an inhibitor of osteoblastogenesis. Stable expression of miR-145 decreased endogenous Cbfb expression and inhibited osteoblastogenesis, in cooperation with miR-34c. Furthermore, miR-145 decreased bone regeneration in vivo. Our results indicate that miR-145 physiologically regulates osteoblast differentiation and bone formation via Cbfb expression by forming a regulatory microRNA network.


Subject(s)
Core Binding Factor beta Subunit/genetics , MicroRNAs/metabolism , Osteoblasts/physiology , 3T3 Cells , Animals , Bone Regeneration , Cell Differentiation , Cell Line , Core Binding Factor beta Subunit/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Mice , Promoter Regions, Genetic
9.
Mol Metab ; 4(3): 175-85, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25737953

ABSTRACT

OBJECTIVE: Bone mass is maintained through a balance of bone formation and resorption. This homeostatic balance is regulated by various systems involving humoral and local factors. The discovery that the anorexigenic hormone leptin regulates bone mass via neuronal pathways revealed that neurons and neuropeptides are intimately involved in bone homeostasis. Ghrelin is a stomach-derived orexigenic hormone that counteracts leptin's action. However, the physiological role of ghrelin in bone homeostasis remains unknown. In this study, through the global knockout of ghrelin receptor (Ghsr) followed by tissue-specific re-expression, we addressed the molecular basis of the action of ghrelin in bone remodeling in vivo. METHODS: We performed molecular, genetic and cell biological analyses of Ghsr-null mice and Ghsr-null mice with tissue specific Ghsr restoration. Furthermore, we evaluated the molecular mechanism of ghrelin by molecular and cell-based assays. RESULTS: Ghsr-null mice showed a low bone mass phenotype with poor bone formation. Restoring the expression of Ghsr specifically in osteoblasts, and not in osteoclasts or the central nervous system, ameliorated bone abnormalities in Ghsr-null mice. Cell-based assays revealed ghrelin induced the phosphorylation of CREB and the expression of Runx2, which in turn accelerated osteoblast differentiation. CONCLUSIONS: Our data show that ghrelin regulates bone remodeling through Ghsr in osteoblasts by modulating the CREB and Runx2 pathways.

10.
Nature ; 497(7450): 490-3, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23644455

ABSTRACT

Semaphorin 3A (Sema3A) is a diffusible axonal chemorepellent that has an important role in axon guidance. Previous studies have demonstrated that Sema3a(-/-) mice have multiple developmental defects due to abnormal neuronal innervations. Here we show in mice that Sema3A is abundantly expressed in bone, and cell-based assays showed that Sema3A affected osteoblast differentiation in a cell-autonomous fashion. Accordingly, Sema3a(-/-) mice had a low bone mass due to decreased bone formation. However, osteoblast-specific Sema3A-deficient mice (Sema3acol1(-/-) and Sema3aosx(-/-) mice) had normal bone mass, even though the expression of Sema3A in bone was substantially decreased. In contrast, mice lacking Sema3A in neurons (Sema3asynapsin(-/-) and Sema3anestin(-/-) mice) had low bone mass, similar to Sema3a(-/-) mice, indicating that neuron-derived Sema3A is responsible for the observed bone abnormalities independent of the local effect of Sema3A in bone. Indeed, the number of sensory innervations of trabecular bone was significantly decreased in Sema3asynapsin(-/-) mice, whereas sympathetic innervations of trabecular bone were unchanged. Moreover, ablating sensory nerves decreased bone mass in wild-type mice, whereas it did not reduce the low bone mass in Sema3anestin(-/-) mice further, supporting the essential role of the sensory nervous system in normal bone homeostasis. Finally, neuronal abnormalities in Sema3a(-/-) mice, such as olfactory development, were identified in Sema3asynasin(-/-) mice, demonstrating that neuron-derived Sema3A contributes to the abnormal neural development seen in Sema3a(-/-) mice, and indicating that Sema3A produced in neurons regulates neural development in an autocrine manner. This study demonstrates that Sema3A regulates bone remodelling indirectly by modulating sensory nerve development, but not directly by acting on osteoblasts.


Subject(s)
Bone Remodeling , Bone and Bones/innervation , Bone and Bones/metabolism , Semaphorin-3A/metabolism , Sensory Receptor Cells/metabolism , Animals , Bone and Bones/anatomy & histology , Cell Differentiation , Cells, Cultured , Female , Male , Mice , Organ Size , Osteoblasts/cytology , Osteoblasts/metabolism , Semaphorin-3A/deficiency , Semaphorin-3A/genetics , Sensory Receptor Cells/cytology
11.
Nat Med ; 18(4): 589-94, 2012 Mar 04.
Article in English | MEDLINE | ID: mdl-22388090

ABSTRACT

Bone homeostasis is maintained by the balance between osteoblastic bone formation and osteoclastic bone resorption. Osteoclasts are multinucleated cells that are formed by mononuclear preosteoclast fusion. Fat-soluble vitamins such as vitamin D are pivotal in maintaining skeletal integrity. However, the role of vitamin E in bone remodeling is unknown. Here, we show that mice deficient in α-tocopherol transfer protein (Ttpa(-/-) mice), a mouse model of genetic vitamin E deficiency, have high bone mass as a result of a decrease in bone resorption. Cell-based assays indicated that α-tocopherol stimulated osteoclast fusion, independent of its antioxidant capacity, by inducing the expression of dendritic-cell-specific transmembrane protein, an essential molecule for osteoclast fusion, through activation of mitogen-activated protein kinase 14 (p38) and microphthalmia-associated transcription factor, as well as its direct recruitment to the Tm7sf4 (a gene encoding DC-STAMP) promoter. Indeed, the bone abnormality seen in Ttpa(-/-) mice was rescued by a Tm7sf4 transgene. Moreover, wild-type mice or rats fed an α-tocopherol-supplemented diet, which contains a comparable amount of α-tocopherol to supplements consumed by many people, lost bone mass. These results show that serum vitamin E is a determinant of bone mass through its regulation of osteoclast fusion.


Subject(s)
Bone Resorption/diet therapy , Bone and Bones/drug effects , Osteoclasts/drug effects , Vitamin E Deficiency/pathology , Vitamin E/administration & dosage , Vitamins/administration & dosage , Amino Acids/blood , Animals , Apoptosis/drug effects , Bone Resorption/etiology , Bone Resorption/genetics , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Bromodeoxyuridine/metabolism , Carrier Proteins/genetics , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chromatin Immunoprecipitation , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , In Situ Nick-End Labeling , Macrophage Colony-Stimulating Factor/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microphthalmia-Associated Transcription Factor/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , Nitrophenols/metabolism , Osteocytes/drug effects , Osteocytes/metabolism , RANK Ligand/metabolism , RNA, Small Interfering/pharmacology , Rats , Signal Transduction/drug effects , Signal Transduction/genetics , Tomography, X-Ray Computed , Transfection , Vitamin E/blood , Vitamin E Deficiency/complications , Vitamin E Deficiency/diet therapy , Vitamin E Deficiency/genetics , Vitamins/blood , alpha-Tocopherol/administration & dosage , alpha-Tocopherol/blood
12.
Proc Natl Acad Sci U S A ; 106(49): 20794-9, 2009 Dec 08.
Article in English | MEDLINE | ID: mdl-19933329

ABSTRACT

Growing evidence shows that microRNAs (miRNAs) regulate various developmental and homeostatic events in vertebrates and invertebrates. Osteoblast differentiation is a key step in proper skeletal development and acquisition of bone mass; however, the physiological role of non-coding small RNAs, especially miRNAs, in osteoblast differentiation remains elusive. Here, through comprehensive analysis of miRNAs expression during osteoblast differentiation, we show that miR-206, previously viewed as a muscle-specific miRNA, is a key regulator of this process. miR-206 was expressed in osteoblasts, and its expression decreased over the course of osteoblast differentiation. Overexpression of miR-206 in osteoblasts inhibited their differentiation, and conversely, knockdown of miR-206 expression promoted osteoblast differentiation. In silico analysis and molecular experiments revealed connexin 43 (Cx43), a major gap junction protein in osteoblasts, as a target of miR-206, and restoration of Cx43 expression in miR-206-expressing osteoblasts rescued them from the inhibitory effect of miR-206 on osteoblast differentiation. Finally, transgenic mice expressing miR-206 in osteoblasts developed a low bone mass phenotype due to impaired osteoblast differentiation. Our data show that miRNA is a regulator of osteoblast differentiation.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation , MicroRNAs/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Animals , Base Sequence , Bone and Bones/metabolism , Bone and Bones/pathology , Cell Line , Connexin 43/genetics , Mice , MicroRNAs/genetics , Molecular Sequence Data , Organ Size , Osteogenesis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...