Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Physiol ; 239(4): e31197, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38284484

RESUMEN

Cytoplasmic polyadenylation element-binding protein 4 (Cpeb4) is an RNA-binding protein that regulates posttranscriptional regulation, such as regulation of messenger RNA stability and translation. In the previous study, we reported that Cpeb4 localizes to nuclear bodies upon induction of osteoclast differentiation by RANKL. However, the mechanisms of the localization of Cpeb4 and osteoclastogenesis by Cpeb4 remain unknown. Here, we show that Cpeb4 localizes to the nuclear bodies by its RNA-binding ability and partially regulates normal splicing during osteoclast differentiation. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis with Phos-tag® revealed that the phosphorylation levels of Cpeb4 were already high in the RAW264.7 cells and were not altered by RANKL treatment. Immunofluorescence showed that exogenous Cpeb4 in HEK293T cells without RANKL stimulation localized to the same foci as shown in RANKL-stimulated RAW264.7 cells. Furthermore, when nuclear export was inhibited by leptomycin B treatment, Cpeb4 accumulated throughout the nucleus. Importantly, RNA recognition motif (RRM) 7 of Cpeb4 was essential for the localization. In contrast, the intrinsically disordered region, RRM1, and zinc finger domain CEBP_ZZ were not necessary for the localization. The mechanistic study showed that Cpeb4 co-localized and interacted with the splicing factors serine/arginine-rich splicing factor 5 (SRSF5) and SRSF6, suggesting that Cpeb4 may be involved in the splicing reaction. RNA-sequencing analysis revealed that the expression of genes related to cell proliferation processes, such as mitotic cell cycle and regulation of cell cycle processes, was elevated in osteoclasts depleted of Cpeb4. Interestingly, the splicing pattern of the inhibitor of DNA binding 2 (Id2) gene, which suppresses osteoclast differentiation, was altered by the depletion of Cpeb4. These results provide new insight into the role of Cpeb4 as a player of normal splicing of Id2 in osteoclast differentiation.


Asunto(s)
Proteína 2 Inhibidora de la Diferenciación , Osteoclastos , Empalme del ARN , Proteínas de Unión al ARN , Humanos , Diferenciación Celular/genética , Células HEK293 , Osteoclastos/citología , Fosfoproteínas/metabolismo , Fosforilación , Ligando RANK/genética , Ligando RANK/farmacología , Ligando RANK/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factores de Empalme Serina-Arginina/metabolismo , Proteína 2 Inhibidora de la Diferenciación/genética , Animales , Ratones
2.
J Cell Physiol ; 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38769895

RESUMEN

Teriparatide is a peptide derived from a parathyroid hormone (PTH) and an osteoporosis therapeutic drug with potent bone formation-promoting activity. To identify novel druggable genes that act downstream of PTH signaling and are potentially involved in bone formation, we screened PTH target genes in mouse osteoblast-like MC3T3-E1 cells. Here we show that Gprc5a, encoding an orphan G protein-coupled receptor, is a novel PTH-inducible gene and negatively regulates osteoblast proliferation and differentiation. PTH treatment induced Gprc5a expression in MC3T3-E1 cells, rat osteosarcoma ROS17/2.8 cells, and mouse femurs. Induction of Gprc5a expression by PTH occurred in the absence of protein synthesis and was mediated primarily via the cAMP pathway, suggesting that Gprc5a is a direct target of PTH signaling. Interestingly, Gprc5a expression was induced additively by co-treatment with PTH and 1α, 25-dihydroxyvitamin D3 (calcitriol), or retinoic acid in MC3T3-E1 cells. Reporter analysis of a 1 kb fragment of human GPRC5A promoter revealed that the promoter fragment showed responsiveness to PTH via the cAMP response element, suggesting that GPRC5A is also a PTH-inducible gene in humans. Gprc5a knockdown promoted cell viability and proliferation, as demonstrated by MTT and BrdU assays. Gprc5a knockdown also promoted osteoblast differentiation, as indicated by gene expression analysis and mineralization assay. Mechanistic studies showed that Gprc5a interacted with BMPR1A and suppressed BMP signaling induced by BMP-2 and constitutively active BMP receptors, ALK2 (ACVR1) Q207D and ALK3 (BMPR1A) Q233D. Thus, our results suggest that Gprc5a is a novel gene induced by PTH that acts in an inhibitory manner on both cell proliferation and osteoblast differentiation and is a candidate for drug targets for osteoporosis.

3.
Biochem Biophys Res Commun ; 719: 150063, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-38749090

RESUMEN

Osteoclasts are multinucleated cells with bone resorption activity. Excessive osteoclast activity has been implicated in osteoporosis, rheumatoid arthritis, and bone destruction due to bone metastases from cancer, making osteoclasts essential target cells in bone and joint diseases. C-terminal domain nuclear envelope phosphatase 1 (Ctdnep1, formerly Dullard) is a negative regulator of transforming growth factor (TGF)-ß superfamily signaling and regulates endochondral ossification in mesenchymal cells during skeletal development. In this study, we investigated the role of Ctdnep1 in the Receptor activator of nuclear factor-kappa B ligand (RANKL)-induced RAW264.7 osteoclast differentiation. Expression of Ctdnep1 did not change during osteoclast differentiation; Ctdnep1 protein localized to the cytoplasm before and after osteoclast differentiation. Small interfering RNA-mediated knockdown of Ctdnep1 increased tartrate-resistant acid phosphatase-positive multinucleated osteoclasts and the expression of osteoclast marker genes, including Acp5, Ctsk, and Nfatc1. Interestingly, the knockdown of Ctdnep1 increased the protein level of Nfatc1 in cells unstimulated with RANKL. Knockdown of Ctdnep1 also enhanced calcium-resorbing activity. Mechanistically, the knockdown of Ctdnep1 increased the phosphorylation of RANKL signaling components. These results suggest that Ctdnep1 negatively regulates osteoclast differentiation by suppressing the RANKL signaling pathway.


Asunto(s)
Diferenciación Celular , Osteoclastos , Ligando RANK , Animales , Ratones , Técnicas de Silenciamiento del Gen , Factores de Transcripción NFATC/metabolismo , Factores de Transcripción NFATC/genética , Osteoclastos/metabolismo , Osteoclastos/citología , Ligando RANK/metabolismo , Células RAW 264.7 , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo
4.
Biochem Biophys Res Commun ; 528(4): 621-627, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32517870

RESUMEN

Cytoplasmic polyadenylation element binding (CPEB) proteins are RNA-binding proteins involved in translational regulation of the specific target mRNAs and control function of various organs including brain, liver and hematopoietic system. However, the role of CPEB proteins during osteoclast differentiation remains unclear. Here we show that Cpeb4 is required for RANKL-induced osteoclast differentiation in mouse macrophage-derived RAW264.7 cell line. Cpeb4 mRNA and protein levels are upregulated at the late stage of osteoclast differentiation. Immunofluorescence analysis revealed that Cpeb4 is translocated from cytoplasm to nuclear bodies in response to RANKL stimulation. Inhibition of PI3K-Akt signaling or calcium-NFAT pathways using chemical inhibitors suppressed nuclear localization of Cpeb4. Loss-of-function analysis showed that shRNA-mediated Cpeb4 depletion strongly impaired TRAP-positive osteoclast formation and expression of key differentiation markers including Acp5, Ctsk, Nfatc1 and Dcstamp. These results suggest that Cpeb4 is a positive regulator in osteoclastogenesis downstream of RANKL signaling.


Asunto(s)
Osteoclastos/citología , Osteogénesis , Proteínas de Unión al ARN/metabolismo , Animales , Ratones , Osteoclastos/metabolismo , Ligando RANK/metabolismo , Células RAW 264.7 , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Transducción de Señal , Regulación hacia Arriba
5.
Genes Cells ; 23(5): 345-356, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29521016

RESUMEN

In mammals, the ovarian follicles are regulated at least in part by bone morphogenetic protein (BMP) family members. Dullard (also known as Ctdnep1) gene encodes a phosphatase that suppresses BMP signaling by inactivating or degrading BMP receptors. Here we report that the Col1a1-Cre-induced Dullard mutant mice displayed hemorrhagic ovarian cysts, with red blood cells accumulated in the follicles, resulting in infertility. Cells expressing Cre driven by Col1a1 2.3-kb promoter and their descendants were found in granulosa cells in the ovary and in Sertoli cells in the testis. DullardmRNA was localized to granulosa cells in the ovary. Genes involved in steroid hormone genesis including Cyp11a1, Hsd3b1 and Star were reduced, whereas expression of Smad6 and Smad7, BMP-inducible inhibitory Smads, was up-regulated in the Dullard mutant ovaries. Tamoxifen-inducible Dullard deletion in the whole body using Rosa26-CreER mice also resulted in hemorrhagic ovarian cysts in 2 weeks, which was rescued by administration of LDN-193189, a chemical inhibitor of BMP receptor kinase, suggesting that the hemorrhage in the Dullard-deficient ovarian follicles might be caused by increased BMP signaling. Thus, we conclude that Dullard is essential for ovarian homeostasis at least in part via suppression of BMP signaling.


Asunto(s)
Colágeno Tipo I/metabolismo , Hemorragia/patología , Infertilidad Femenina/patología , Quistes Ováricos/patología , Folículo Ovárico/patología , Fosfoproteínas Fosfatasas/deficiencia , Animales , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Huesos/metabolismo , Huesos/patología , Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hemorragia/metabolismo , Infertilidad Femenina/metabolismo , Masculino , Ratones , Ratones Noqueados , Quistes Ováricos/metabolismo , Folículo Ovárico/metabolismo , Fosfoproteínas Fosfatasas/fisiología , Fosforilación , Pirazoles/farmacología , Pirimidinas/farmacología , Transducción de Señal , Proteínas Smad/metabolismo , Testículo/metabolismo , Testículo/patología
6.
Biochem Biophys Res Commun ; 498(4): 967-974, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29548825

RESUMEN

Bone mass is determined by coordinated acts of osteoblasts and osteoclasts, which control bone formation and resorption, respectively. Osteoclasts are multinucleated, macrophage/monocyte lineage cells from bone marrow. The Dok-family adaptors Dok-1, Dok-2 and Dok-3 are expressed in the macrophage/monocyte lineage and negatively regulate many signaling pathways, implying roles in osteoclastogenesis. Indeed, mice lacking Dok-1 and Dok-2, the closest homologues with redundant functions, develop osteopenia with increased osteoclast counts compared to the wild-type controls. Here, we demonstrate that Dok-3 knockout (KO) mice also develop osteopenia. However, Dok-3 KO, but not Dok-1/-2 double-KO (DKO), mice develop larger osteoclasts within the normal cell-count range, suggesting a distinctive role for Dok-3. Indeed, Dok-3 KO, but not Dok-1/-2 DKO, bone marrow-derived cells (BMDCs) generated larger osteoclasts with more nuclei due to augmented cell-to-cell fusion in vitro. In addition, while Dok-1/-2 DKO BMDCs generated more osteoclasts, Dok-1/-2/-3 triple-KO (TKO) BMDCs generated osteoclasts increased in both number and size. Furthermore, Dok-1/-2/-3 TKO mice showed the combined effects of Dok-3 and Dok-1/-2 deficiency: severe osteopenia with more and larger osteoclasts. Together, our findings demonstrate that Dok-3 and Dok-1/-2 play distinctive but cooperative roles in osteoclastogenesis and protect mice from osteopenia, providing physiological and pathophysiological insight into bone homeostasis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Enfermedades Óseas Metabólicas/prevención & control , Proteínas de Unión al ADN/fisiología , Osteoclastos/citología , Osteogénesis , Fosfoproteínas/fisiología , Proteínas de Unión al ARN/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células de la Médula Ósea/citología , Recuento de Células , Técnicas de Cultivo de Célula , Fusión Celular , Proliferación Celular , Tamaño de la Célula , Proteínas de Unión al ADN/genética , Ratones , Ratones Noqueados , Fosfoproteínas/genética , Proteínas de Unión al ARN/genética
7.
Proc Natl Acad Sci U S A ; 112(50): 15432-7, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26621720

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Huesos/citología , Movimiento Celular , Proteínas Oncogénicas/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Animales , Resorción Ósea/metabolismo , Resorción Ósea/patología , Huesos/diagnóstico por imagen , Huesos/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Células Cultivadas , Técnicas de Silenciamiento del Gen , Proteínas Sustrato del Receptor de Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Ratones Noqueados , Proteínas Oncogénicas/deficiencia , Tamaño de los Órganos , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Radiografía , Cráneo/efectos de los fármacos , Cráneo/metabolismo , Cicatrización de Heridas/efectos de los fármacos
8.
Proc Natl Acad Sci U S A ; 111(7): 2692-7, 2014 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-24550297

RESUMEN

Osteoclastogenesis is under the control of posttranscriptional and transcriptional events. However, posttranscriptional regulation of osteoclastogenesis is incompletely understood. CNOT3 is a component of the CCR4 family that regulates mRNA stability, but its function in bone is not known. Here, we show that Cnot3 deficiency by deletion of a single allele induces osteoporosis. Cnot3 deficiency causes an enhancement in bone resorption in association with an elevation in bone formation, resulting in high-turnover type bone loss. At the cellular level, Cnot3 deficiency enhances receptor activator of NF-κB ligand (RANKL) effects on osteoclastogenesis in a cell-autonomous manner. Conversely, Cnot3 deficiency does not affect osteoblasts directly. Cnot3 deficiency does not alter RANKL expression but enhances receptor activator of NF-κB (RANK) mRNA expression in bone in vivo. Cnot3 deficiency promotes RANK mRNA stability about twofold in bone marrow cells of mice. Cnot3 knockdown also increases RANK mRNA expression in the precursor cell line for osteoclasts. Anti-CNOT3 antibody immunoprecipitates RANK mRNA. Cnot3 deficiency stabilizes luciferase reporter expression linked to the 3'-UTR fragment of RANK mRNA. In contrast, Cnot3 overexpression destabilizes the luciferase reporter linked to RANK 3'-UTR. In aged mice that exhibit severe osteoporosis, Cnot3 expression levels in bone are reduced about threefold in vivo. Surprisingly, Cnot3 deficiency in these aged mice further exacerbates osteoporosis, which also occurs via enhancement of osteoclastic activity. Our results reveal that CNOT3 is a critical regulator of bone mass acting on bone resorption through posttranscriptional down-regulation of RANK mRNA stability, at least in part, even in aging-induced osteoporosis.


Asunto(s)
Resorción Ósea/fisiopatología , Regulación de la Expresión Génica/fisiología , Osteoporosis/fisiopatología , Estabilidad del ARN/fisiología , ARN Mensajero/metabolismo , Factores de Transcripción/metabolismo , Absorciometría de Fotón , Factores de Edad , Animales , Densidad Ósea , Cartilla de ADN/genética , Imagenología Tridimensional , Luciferasas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estabilidad del ARN/genética , ARN Interferente Pequeño/genética , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Microtomografía por Rayos X
9.
J Cell Biochem ; 117(4): 970-7, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26378628

RESUMEN

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.


Asunto(s)
Artritis Experimental/genética , Cartílago Articular/inmunología , Metaloproteinasa 3 de la Matriz/inmunología , Proteínas Asociadas a Matriz Nuclear/inmunología , Ligando RANK/inmunología , Factores de Transcripción/inmunología , Animales , Artritis Experimental/inducido químicamente , Artritis Experimental/inmunología , Artritis Experimental/patología , Autoanticuerpos/biosíntesis , Resorción Ósea , Cartílago Articular/patología , Condrocitos/inmunología , Condrocitos/patología , Femenino , Regulación de la Expresión Génica , Glucosa-6-Fosfato Isomerasa/antagonistas & inhibidores , Glucosa-6-Fosfato Isomerasa/genética , Glucosa-6-Fosfato Isomerasa/inmunología , Sueros Inmunes/administración & dosificación , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Articulaciones/inmunología , Articulaciones/patología , Masculino , Metaloproteinasa 3 de la Matriz/genética , Ratones , Ratones Noqueados , Proteínas Asociadas a Matriz Nuclear/deficiencia , Proteínas Asociadas a Matriz Nuclear/genética , Regiones Promotoras Genéticas , Ligando RANK/genética , Índice de Severidad de la Enfermedad , Transducción de Señal , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Transcripción Genética
10.
J Cell Physiol ; 230(11): 2788-95, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25828538

RESUMEN

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.


Asunto(s)
Cilios/metabolismo , Osteoartritis/genética , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Animales , Cartílago Articular/metabolismo , Condrocitos/metabolismo , Cilios/patología , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Osteoartritis/metabolismo , Osteoartritis/patología , Fosforilación , Transducción de Señal , Factor de Crecimiento Transformador beta/administración & dosificación , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
11.
J Cell Biochem ; 116(1): 142-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25164990

RESUMEN

As the aged population is soaring, prevalence of osteoporosis is increasing. However, the molecular basis underlying the regulation of bone mass is still incompletely understood. Sympathetic tone acts via beta2 adrenergic receptors in bone and regulates the mass of bone which is the target organ of parathyroid hormone (PTH). However, whether beta2 adrenergic receptor is regulated by PTH in bone cells is not known. We therefore investigated the effects of PTH on beta2 adrenergic receptor gene expression in osteoblast-like MC3T3-E1 cells. PTH treatment immediately suppressed the expression levels of beta2 adrenergic receptor mRNA. This PTH effect was dose-dependent starting as low as 1 nM. PTH action on beta2 adrenergic receptor gene expression was inhibited by a transcriptional inhibitor, DRB, but not by a protein synthesis inhibitor, cycloheximide suggesting direct transcription control. Knockdown of beta2 adrenergic receptor promoted PTH-induced expression of c-fos, an immediate early response gene. With respect to molecular basis for this phenomenon, knockdown of beta2 adrenergic receptor enhanced PTH-induced transcriptional activity of cyclic AMP response element-luciferase construct in osteoblasts. Knockdown of beta2 adrenergic receptors also enhanced forskolin-induced luciferase expression, revealing that adenylate cyclase activity is influenced by beta2 adrenergic receptor. As for phosphorylation of transcription factor, knockdown of beta2 adrenergic receptor enhanced PTH-induced phosphorylation of cyclic AMP response element binding protein (CREB). These data reveal that beta2 adrenergic receptor is one of the targets of PTH and acts as a suppressor of PTH action in osteoblasts.


Asunto(s)
Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Hormona Paratiroidea/farmacología , Receptores Adrenérgicos beta 2/metabolismo , Animales , Línea Celular , Colforsina/farmacología , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transducción de Señal/efectos de los fármacos
12.
J Cell Biochem ; 116(6): 1144-52, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25536656

RESUMEN

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.


Asunto(s)
Fosfatasa Alcalina/metabolismo , Proteínas Morfogenéticas Óseas/farmacología , Osteoblastos/metabolismo , Receptores Adrenérgicos/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular , Ratones , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
13.
J Bone Miner Metab ; 33(1): 48-54, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24831120

RESUMEN

Rodent hindlimb unloading (HU) by tail-suspension is a model to investigate disuse-induced bone loss in vivo. Previously, we have shown that osteopontin (OPN, also known as Spp1) is required for unloading-induced bone loss. However, how unloading affects OPN expression in the body is not fully understood. Here, we examined OPN expression in peripheral blood of mice subjected to HU. Real-time RT-PCR analysis indicated that OPN expression is increased in circulating peripheral blood cells. This HU-induced increase in OPN mRNA expression was specific in circulating peripheral blood cells, as OPN was not increased in the blood cells in bone marrow. HU-induced enhancement in OPN expression in peripheral blood cells was associated with an increase in the fraction of monocyte/macrophage lineage cells in the peripheral blood. In contrast, HU decreased the fraction size of B-lymphocytes in the peripheral blood. We further examined if B-lymphogenesis is affected in the mice deficient for osteopontin subjected to HU. In bone marrow, HU decreased the population of the B-lymphocyte lineage cells significantly, whereas it did not alter the population of monocyte/macrophage lineage cells. HU also increased the cells in T-lymphocyte lineage in bone marrow. Interestingly, these changes were observed similarly both in OPN-deficient and wild-type mice. These results indicate for the first time that HU increases OPN expression in circulating cells and suppresses bone marrow B-lymphogenesis.


Asunto(s)
Linfocitos B/citología , Células de la Médula Ósea/citología , Suspensión Trasera , Osteopontina/sangre , Animales , Médula Ósea , Resorción Ósea/metabolismo , Huesos/metabolismo , Linaje de la Célula , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Imagenología Tridimensional , Leucocitos Mononucleares/citología , Macrófagos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/citología , Osteoclastos/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal , Microtomografía por Rayos X
14.
Proc Natl Acad Sci U S A ; 109(19): 7433-8, 2012 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-22538810

RESUMEN

Parathyroid hormone (PTH), the major calcium-regulating hormone, and norepinephrine (NE), the principal neurotransmitter of sympathetic nerves, regulate bone remodeling by activating distinct cell-surface G protein-coupled receptors in osteoblasts: the parathyroid hormone type 1 receptor (PTHR) and the ß(2)-adrenergic receptor (ß(2)AR), respectively. These receptors activate a common cAMP/PKA signal transduction pathway mediated through the stimulatory heterotrimeric G protein. Activation of ß(2)AR via the sympathetic nervous system decreases bone formation and increases bone resorption. Conversely, daily injection of PTH (1-34), a regimen known as intermittent (i)PTH treatment, increases bone mass through the stimulation of trabecular and cortical bone formation and decreases fracture incidences in severe cases of osteoporosis. Here, we show that iPTH has no osteoanabolic activity in mice lacking the ß(2)AR. ß(2)AR deficiency suppressed both iPTH-induced increase in bone formation and resorption. We showed that the lack of ß(2)AR blocks expression of iPTH-target genes involved in bone formation and resorption that are regulated by the cAMP/PKA pathway. These data implicate an unexpected functional interaction between PTHR and ß(2)AR, two G protein-coupled receptors from distinct families, which control bone formation and PTH anabolism.


Asunto(s)
Huesos/efectos de los fármacos , Hormona Paratiroidea/farmacología , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Absorciometría de Fotón , Anabolizantes/metabolismo , Anabolizantes/farmacología , Animales , Densidad Ósea/efectos de los fármacos , Huesos/diagnóstico por imagen , Huesos/metabolismo , Femenino , Fémur/efectos de los fármacos , Fémur/metabolismo , Fluoresceínas , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Hormona Paratiroidea/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/genética , Receptores Adrenérgicos beta 2/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Microtomografía por Rayos X
15.
J Cell Physiol ; 229(10): 1353-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24604668

RESUMEN

Bone metabolism is maintained via balanced repetition of bone resorption by osteoclasts and bone formation by osteoblasts. Osteoblastic cells are capable of conducting self-renewal and differentiation that are basically associated with cell-cycle transition to enable cell specification and bone formation. Osteoblasts are also migrating to fill the resorption cavity curved by osteoclasts during bone remodeling to maintain homeostasis of bone mass whose imbalance leads to osteoporosis. However, technical difficulties have hampered the research on the dynamic relationship between cell cycle and migration in osteoblasts. In this report, we overcome these problems by introducing fluorescent ubiquitination-based cell cycle indicator (FUCCI) reporter system in calvarial osteoblastic cells and reveal that the cells in G1 as well as S/G2 /M phase are migrating. Furthermore, the osteoblastic cells in S/G2 /M phase migrate faster than those in G1 phase. Interestingly, parathyroid hormone (PTH) as an anabolic agent enhances migration velocity of the cells. Mechanical stress, another anabolic signal, also enhances migration velocity. In contrast, in the presence of both PTH and mechanical stress, the migration velocity returns to the base line levels revealing the interaction between the two anabolic stimuli in the regulation of cell migration. Importantly, PTH and mechanical stress also interact when they regulate the transition of cell cycle. These data demonstrate that osteoblastic migration is linked to cell cycle and it is under the control of mechanical and chemical stimuli that coordinate to regulate bone mass.


Asunto(s)
Técnicas Biosensibles , Remodelación Ósea , Ciclo Celular , Movimiento Celular , Rastreo Celular/métodos , Mecanotransducción Celular , Osteoblastos/metabolismo , Hormona Paratiroidea/metabolismo , Animales , Células Cultivadas , Genes Reporteros , Proteínas Luminiscentes/biosíntesis , Proteínas Luminiscentes/genética , Ratones , Ratones Transgénicos , Estrés Mecánico , Factores de Tiempo
16.
Proc Natl Acad Sci U S A ; 108(43): 17767-72, 2011 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-21990347

RESUMEN

The sympathetic nervous system suppresses bone mass by mechanisms that remain incompletely elucidated. Using cell-based and murine genetics approaches, we show that this activity of the sympathetic nervous system requires osteopontin (OPN), a cytokine and one of the major members of the noncollagenous extracellular matrix proteins of bone. In this work, we found that the stimulation of the sympathetic tone by isoproterenol increased the level of OPN expression in the plasma and bone and that mice lacking OPN (OPN-KO) suppressed the isoproterenol-induced bone loss by preventing reduced osteoblastic and enhanced osteoclastic activities. In addition, we found that OPN is necessary for changes in the expression of genes related to bone resorption and bone formation that are induced by activation of the sympathetic tone. At the cellular level, we showed that intracellular OPN modulated the capacity of the ß2-adrenergic receptor to generate cAMP with a corresponding modulation of cAMP-response element binding (CREB) phosphorylation and associated transcriptional events inside the cell. Our results indicate that OPN plays a critical role in sympathetic tone regulation of bone mass and that this OPN regulation is taking place through modulation of the ß2-adrenergic receptor/cAMP signaling system.


Asunto(s)
Huesos/fisiología , Osteopontina/metabolismo , Sistema Nervioso Simpático/fisiología , Análisis de Varianza , Animales , Huesos/metabolismo , AMP Cíclico/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Isoproterenol/farmacología , Ratones , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteopontina/deficiencia , Receptores Adrenérgicos beta 2/metabolismo , Sistema Nervioso Simpático/efectos de los fármacos
17.
Front Cell Dev Biol ; 12: 1370723, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38989059

RESUMEN

Juvenile nephronophthisis is an inherited renal ciliopathy with cystic kidney disease, renal fibrosis, and end-stage renal failure in children and young adults. Mutations in the NPHP1 gene encoding nephrocystin-1 protein have been identified as the most frequently responsible gene and cause the formation of cysts in the renal medulla. The molecular pathogenesis of juvenile nephronophthisis remains elusive, and no effective medicines to prevent end-stage renal failure exist even today. No human cellular models have been available yet. Here, we report a first disease model of juvenile nephronophthisis using patient-derived and gene-edited human induced pluripotent stem cells (hiPSCs) and kidney organoids derived from these hiPSCs. We established NPHP1-overexpressing hiPSCs from patient-derived hiPSCs and NPHP1-deficient hiPSCs from healthy donor hiPSCs. Comparing these series of hiPSCs, we found abnormalities in primary cilia associated with NPHP1 deficiency in hiPSCs. Kidney organoids generated from the hiPSCs lacking NPHP1 formed renal cysts frequently in suspension culture with constant rotation. This cyst formation in patient-derived kidney organoids was rescued by overexpression of NPHP1. Transcriptome analysis on these kidney organoids revealed that loss of NPHP1 caused lower expression of genes related to primary cilia in epithelial cells and higher expression of genes related to the cell cycle. These findings suggested the relationship between abnormality in primary cilia induced by NPHP1 loss and abnormal proliferative characteristics in the formation of renal cysts. These findings demonstrated that hiPSC-based systematic disease modeling of juvenile nephronophthisis contributed to elucidating the molecular pathogenesis and developing new therapies.

18.
Regen Ther ; 26: 71-79, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38828011

RESUMEN

Background: Human dental pulp-derived stem cells (hDPSCs) have emerged as a promising source for adult stem cell-based regenerative medicine. Stage-specific embryonic antigen 3 (SSEA3) is a cell surface marker associated with Multilineage-differentiating stress-enduring (Muse) cells, a subpopulation of human bone marrow-derived stem cells (hBMSCs), known for their potent regenerative potential and safety profile. In this study, we investigated the influence of the prolonged culture period and the number of culture passages on the regenerative capacity of hDPSCs and explored the association between SSEA3 expression and their regenerative abilities. Methods: hDPSCs were isolated and cultured for up to 20 passages. Cell proliferation, migration, and osteogenic, adipogenic and neurogenic differentiation potential were assessed at passages 5, 10, and 20. Flow cytometry and immunofluorescence were employed to analyze SSEA3 expression. RNA sequencing (RNA-seq) was performed on SSEA3-positive and SSEA3-negative hDPSCs to identify differentially expressed genes and associated pathways. Results: Our findings demonstrated a progressive decline in hDPSCs proliferation and migration capacity with increasing passage number. Conversely, cell size exhibited a positive correlation with passage number. Early passage hDPSCs displayed superior osteogenic and adipogenic differentiation potential. Notably, SSEA3 expression exhibited a significant negative correlation with passage numbers, reflecting the observed decline in differentiation capacity. RNA-seq analysis revealed distinct transcriptional profiles between SSEA3-positive and SSEA3-negative hDPSCs. SSEA3-positive cells displayed upregulation of genes associated with ectodermal differentiation and downregulation of genes involved in cell adhesion. Conclusions: This study elucidates the impact of passaging on hDPSC behavior and suggests SSEA3 as a valuable biomarker for evaluating stemness and regenerative potential. SSEA3-positive hDPSCs, functionally analogous to Muse cells, represent a promising cell population for developing targeted regenerative therapies with potentially improved clinical outcomes.

19.
Stem Cell Res ; 79: 103493, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-39032428

RESUMEN

Myelin basic protein (MBP) is a major component of the myelin sheaths of oligodendrocytes in the central nervous system and Schwann cells of the peripheral nervous system. Here we generated heterozygous fluorescent reporter of MBP gene in human induced pluripotent stem cells (hiPSCs). CRISPR/Cas9 genome editing technology was employed to knock in fused tdTomato fluorescent protein and EF1 alpha promoter-driven Bleomycin (Zeocin) resistance gene to the translational MBP C-terminal region. The resulting line, MBP-TEZ, showed tdTomato fluorescence upon oligodendrocyte differentiation. This reporter hiPSC line provides a precedential opportunity for monitoring human myelin formation and degeneration and purifying MBP-expressing cell lineages.

20.
J Biol Chem ; 287(40): 33545-53, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22773831

RESUMEN

Bone development is a dynamic process that requires cell motility and morphological adaptation under the control of actin cytoskeleton. This actin cytoskeleton system is regulated by critical modulators including actin-binding proteins. Among them, profilin1 (Pfn1) is a key player to control actin fiber structure, and it is involved in a number of cellular activities such as migration. During the early phase of body development, skeletal stem cells and osteoblastic progenitor cells migrate to form initial rudiments for future skeletons. During this migration, these cells extend their process based on actin cytoskeletal rearrangement to locate themselves in an appropriate location within microenvironment. However, the role of Pfn1 in regulation of mesenchymal progenitor cells (MPCs) during skeletal development is incompletely understood. Here we examined the role of Pfn1 in skeletal development using a genetic ablation of Pfn1 in MPCs by using Prx1-Cre recombinase. We found that Pfn1 deficiency in MPCs caused complete cleft sternum. Notably, Pfn1-deficient mice exhibited an absence of trabecular bone in the marrow space of appendicular long bone. This phenotype is location-specific, as Pfn1 deficiency did not largely affect osteoblasts in cortical bone. Pfn1 deficiency also suppressed longitudinal growth of long bone. In vitro, Pfn1 deficiency induced retardation of osteoblastic cell migration. These observations revealed that Pfn1 is a critical molecule for the skeletal development, and this could be at least in part associated with the retardation of cell migration.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Profilinas/fisiología , Alelos , Animales , Huesos/metabolismo , Cartílago/metabolismo , Movimiento Celular , Citoesqueleto/metabolismo , Genotipo , Células Madre Mesenquimatosas/citología , Ratones , Ratones Noqueados , Ratones Transgénicos , Células 3T3 NIH , Osteoblastos/citología , Osteogénesis , Profilinas/metabolismo , ARN Interferente Pequeño/metabolismo , Factores de Tiempo , Transfección , Microtomografía por Rayos X/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA