Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Int J Mol Sci ; 24(17)2023 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-37686441

RESUMEN

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.


Asunto(s)
Osteocitos , Osteogénesis , ARN Largo no Codificante , Animales , Ratones , Cromatografía Liquida , Osteogénesis/genética , ARN Largo no Codificante/genética , Espectrometría de Masas en Tándem , Vía de Señalización Wnt
2.
Proc Natl Acad Sci U S A ; 115(9): 2204-2209, 2018 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-29440427

RESUMEN

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.


Asunto(s)
Neoplasias Óseas/metabolismo , Neoplasias de la Mama/patología , Proteínas Activadoras de GTPasa/metabolismo , Proteínas de la Membrana/metabolismo , MicroARNs/metabolismo , Neoplasias de la Próstata/metabolismo , Adenocarcinoma/metabolismo , Animales , Neoplasias Óseas/secundario , Sustitutos de Huesos , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Proteínas Activadoras de GTPasa/genética , Humanos , Masculino , Proteínas de la Membrana/genética , Células Madre Mesenquimatosas , Ratones , MicroARNs/genética , Neoplasias Experimentales/metabolismo
3.
Nature ; 497(7450): 490-3, 2013 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-23644455

RESUMEN

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.


Asunto(s)
Remodelación Ósea , Huesos/inervación , Huesos/metabolismo , Semaforina-3A/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Huesos/anatomía & histología , Diferenciación Celular , Células Cultivadas , Femenino , Masculino , Ratones , Tamaño de los Órganos , Osteoblastos/citología , Osteoblastos/metabolismo , Semaforina-3A/deficiencia , Semaforina-3A/genética , Células Receptoras Sensoriales/citología
4.
Int J Mol Sci ; 20(5)2019 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-30832329

RESUMEN

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.


Asunto(s)
Condrogénesis , Curación de Fractura , Osteogénesis , Pericitos/citología , Cultivo Primario de Células/métodos , Animales , Diferenciación Celular , Línea Celular , Células Cultivadas , Condrocitos/citología , Trasplante de Células Madre Mesenquimatosas/métodos , Ratones , Ratones Endogámicos BALB C , Osteoblastos/citología , Pericitos/trasplante
5.
J Infect Dis ; 215(12): 1893-1897, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28525596

RESUMEN

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.


Asunto(s)
Terapia Antirretroviral Altamente Activa/efectos adversos , Densidad Ósea/efectos de los fármacos , Inhibidores de Proteasas/efectos adversos , Animales , Arginina/análogos & derivados , Arginina/sangre , Biomarcadores/sangre , Femenino , VIH-1/efectos de los fármacos , Humanos , Inhibidores de Integrasa , Lisina/análogos & derivados , Lisina/sangre , Masculino , Ratones , Osteocalcina/sangre , Estudios Retrospectivos , Inhibidores de la Transcriptasa Inversa
6.
Sci Rep ; 13(1): 4674, 2023 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-36949102

RESUMEN

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.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina , Imagenología Tridimensional , Ratones , Humanos , Animales , Huesos/diagnóstico por imagen , Remodelación Ósea , Redes Neurales de la Computación
7.
Proc Natl Acad Sci U S A ; 106(49): 20794-9, 2009 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-19933329

RESUMEN

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.


Asunto(s)
Diferenciación Celular/genética , Regulación de la Expresión Génica , MicroARNs/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Animales , Secuencia de Bases , Huesos/metabolismo , Huesos/patología , Línea Celular , Conexina 43/genética , Ratones , MicroARNs/genética , Datos de Secuencia Molecular , Tamaño de los Órganos , Osteogénesis/genética
8.
J Bone Miner Res ; 31(7): 1344-55, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26841172

RESUMEN

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.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Resorción Ósea/metabolismo , Relojes Circadianos/fisiología , Osteoclastos/metabolismo , Factores de Transcripción ARNTL/genética , Animales , Resorción Ósea/genética , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Diferenciación Celular/fisiología , Ratones , Ratones Noqueados , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
9.
JCI Insight ; 1(5): e85061, 2016 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-27699258

RESUMEN

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.


Asunto(s)
Calcineurina/metabolismo , Inhibidores de Histona Desacetilasas/uso terapéutico , Ácidos Hidroxámicos/uso terapéutico , Indoles/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones Endogámicos NOD , Ratones SCID , Panobinostat , Tacrolimus/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Mol Metab ; 4(3): 175-85, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25737953

RESUMEN

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.

11.
FEBS Lett ; 589(21): 3302-8, 2015 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-26450370

RESUMEN

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.


Asunto(s)
Subunidad beta del Factor de Unión al Sitio Principal/genética , MicroARNs/metabolismo , Osteoblastos/fisiología , Células 3T3 , Animales , Regeneración Ósea , Diferenciación Celular , Línea Celular , Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Ratones , Regiones Promotoras Genéticas
12.
Nat Med ; 18(4): 589-94, 2012 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-22388090

RESUMEN

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.


Asunto(s)
Resorción Ósea/dietoterapia , Huesos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Deficiencia de Vitamina E/patología , Vitamina E/administración & dosificación , Vitaminas/administración & dosificación , Aminoácidos/sangre , Animales , Apoptosis/efectos de los fármacos , Resorción Ósea/etiología , Resorción Ósea/genética , Huesos/diagnóstico por imagen , Huesos/patología , Bromodesoxiuridina/metabolismo , Proteínas Portadoras/genética , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Inmunoprecipitación de Cromatina , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Etiquetado Corte-Fin in Situ , Factor Estimulante de Colonias de Macrófagos/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción Asociado a Microftalmía/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Nitrofenoles/metabolismo , Osteocitos/efectos de los fármacos , Osteocitos/metabolismo , Ligando RANK/metabolismo , ARN Interferente Pequeño/farmacología , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tomografía Computarizada por Rayos X , Transfección , Vitamina E/sangre , Deficiencia de Vitamina E/complicaciones , Deficiencia de Vitamina E/dietoterapia , Deficiencia de Vitamina E/genética , Vitaminas/sangre , alfa-Tocoferol/administración & dosificación , alfa-Tocoferol/sangre
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda