Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Biochem Funct ; 42(4): e4064, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38807466

RESUMEN

Human dental pulp stem cells are a potentially useful resource for cell-based therapies and tissue repair in dental and medical applications. However, the primary culture of isolated dental pulp stem cells has notably been limited. A major requirement of an ideal human dental pulp stem cell culture system is the preservation of efficient proliferation and innate stemness over prolonged passaging, while also ensuring ease of handling through standard, user-friendly culture methods. In this study, we have engineered a novel human dental pulp stem cell line, distinguished by the constitutive expression of telomerase reverse transcriptase (TERT), and the conditional expression of the R24C mutant cyclin-dependent kinase 4 (CDK4R24C) and Cyclin D1. We have named this cell line Tet-off K4DT hDPSCs. Furthermore, we have conducted a comprehensive comparative analysis of their biological attributes in relation to a previously immortalized human dental pulp stem cells, hDPSC-K4DT, which were immortalized by the constitutive expression of CDK4R24C, Cyclin D1 and TERT. In Tet-off K4DT cells, the expression of the K4D genes can be precisely suppressed by the inclusion of doxycycline. Remarkably, Tet-off K4DT cells demonstrated an extended cellular lifespan, increased proliferative capacity, and enhanced osteogenic differentiation potential when compared to K4DT cells. Moreover, Tet-off K4DT cells had no observable genomic aberrations and also displayed a sustained expression of stem cell markers even at relatively advanced passages. Taken together, the establishment of this new cell line holds immense promise as powerful experimental tool for both fundamental and applied research involving dental pulp stem cells.


Asunto(s)
Proliferación Celular , Quinasa 4 Dependiente de la Ciclina , Pulpa Dental , Doxiciclina , Células Madre , Humanos , Pulpa Dental/citología , Pulpa Dental/metabolismo , Proliferación Celular/efectos de los fármacos , Doxiciclina/farmacología , Células Madre/metabolismo , Células Madre/citología , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/genética , Telomerasa/metabolismo , Telomerasa/genética , Ciclina D1/metabolismo , Ciclina D1/genética , Diferenciación Celular/efectos de los fármacos , Células Cultivadas
2.
Connect Tissue Res ; 65(3): 237-252, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38739041

RESUMEN

PURPOSE/AIM OF STUDY: During the development of the vertebrate skeleton, the progressive differentiation and maturation of chondrocytes from mesenchymal progenitors is precisely coordinated by multiple secreted factors and signaling pathways. The WNT signaling pathway has been demonstrated to play a major role in chondrogenesis. However, the identification of secreted factors that fine-tune WNT activity has remained elusive. Here, in this study, we have identified PI15 (peptidase inhibitor 15, protease Inhibitor 15, SugarCrisp), a member of the CAP (cysteine rich secretory proteins, antigen 5, and pathogenesis related 1 proteins) protein superfamily, as a novel secreted WNT antagonist dynamically upregulated during chondrocyte differentiation. MATERIALS AND METHODS: ATDC5 cells, C3H10T1/2 micromass cultures and primary chondrocyte cells were used as in vitro models of chondrogenesis. PI15 levels were stably depleted or overexpressed by viral shRNA or expression vectors. Chondrogenesis was evaluated by qPCR gene expression analysis and Alcian blue staining. Protein interactions were determined by coimmunoprecipitation assays. RESULTS AND CONCLUSIONS: shRNA-mediated knockdown of PI15 in ATDC5 cells, C3H10T1/2 cells or primary chondrocytes inhibits chondrogenesis, whereas the overexpression of PI15 strongly enhances chondrogenic potential. Mechanistically, PI15 binds to the LRP6 WNT co-receptor and blocks WNT-induced LRP6 phosphorylation, thus repressing WNT-induced transcriptional activity and alleviating the inhibitory effect of WNT signaling on chondrogenesis. Altogether, our findings suggest that PI15 acts as a key regulator of chondrogenesis and unveils a mechanism through which chondrocyte-derived molecules can modulate WNT activity as differentiation proceeds, thereby creating a positive feedback loop that further drives differentiation.


Asunto(s)
Diferenciación Celular , Condrocitos , Condrogénesis , Vía de Señalización Wnt , Condrocitos/metabolismo , Condrocitos/efectos de los fármacos , Condrocitos/citología , Diferenciación Celular/efectos de los fármacos , Animales , Vía de Señalización Wnt/efectos de los fármacos , Ratones , Condrogénesis/efectos de los fármacos , Línea Celular , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo
3.
FASEB Bioadv ; 5(2): 62-70, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36816515

RESUMEN

The NF-κB family of transcription factors plays an important role in skeletal development and bone homeostasis. In osteoblast cells, NF-κB signaling has been shown to suppress survival, proliferation, and differentiation. Furthermore, pharmacological suppression of NF-κB enhances osteoblast differentiation and bone formation. Thus, NF-κB antagonists are promising candidates as anabolic agents for enhancing bone mass. In this study, we describe the mechanism by which nobiletin, an inhibitor of NF-κB activity, regulates osteoblast differentiation and mineralization. We found that in MC3T3-E1 osteoblast cells, nobiletin inhibited a TNF-α responsive NF-κB luciferase reporter and also decreased the induction of classical NF-κB target genes by TNF-α. Consistent with this, nobiletin prevented TNF-α -mediated suppression of osteogenesis and potently enhanced the differentiation and mineralization of MC3T3-E1 cells. Likewise, in an in vivo BMP2-induced ectopic bone formation assay, nobiletin markedly enhanced ossicle bone volume. Western blotting and SMAD-responsive luciferase assays also demonstrated that NF-κB suppression of BMP signaling could be inhibited by nobiletin. Thus, our data suggest that mechanistically, nobiletin prevents the endogenous repression of BMP signaling by TNF-α, thereby enhancing osteoblast activity. In conclusion, nobiletin is a novel NF-κB antagonist that may be a useful anabolic agent for bone formation.

4.
BMC Cancer ; 22(1): 936, 2022 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-36038818

RESUMEN

BACKGROUND: Melanoma is a malignant tumor characterized by high proliferation and aggressive metastasis. To address the molecular mechanisms of the proto-oncogene, Rous sarcoma oncogene (Src), which is highly activated and promotes cell proliferation, migration, adhesion, and metastasis in melanoma. Plectin, a cytoskeletal protein, has recently been identified as a Src-binding protein that regulates Src activity in osteoclasts. Plectin is a candidate biomarker of certain tumors because of its high expression and the target of anti-tumor reagents such as ruthenium pyridinecarbothioamide. The molecular mechanisms by which plectin affects melanoma is still unclear. In this study, we examined the role of plectin in melanoma tumor formation. METHODS: We used CRISPR/Cas9 gene editing to knock-out plectin in B16 mouse melanoma cells. Protein levels of plectin and Src activity were examined by western blotting analysis. In vivo tumor formation was assessed by subcutaneous injection of B16 cells into nude mice and histological analysis performed after 2 weeks by Hematoxylin-Eosin (H&E) staining. Cell proliferation was evaluated by direct cell count, cell counting kit-8 assays, cyclin D1 mRNA expression and Ki-67 immunostaining. Cell aggregation and adhesion were examined by spheroid formation, dispase-based dissociation assay and cell adhesion assays. RESULTS: In in vivo tumor formation assays, depletion of plectin resulted in low-density tumors with large intercellular spaces. In vitro experiments revealed that plectin-deficient B16 cells exhibit reduced cell proliferation and reduced cell-to-cell adhesion. Since Src activity is reduced in plectin-deficient melanomas, we examined the relationship between plectin and Src signaling. Src overexpression in plectin knockout B16 cells rescued cell proliferation and improved cell-to-cell adhesion and cell to extracellular matrix adhesion. CONCLUSION: These results suggest that plectin plays critical roles in tumor formation by promoting cell proliferation and cell-to-cell adhesion through Src signaling activity in melanoma cells.


Asunto(s)
Melanoma Experimental , Sarcoma Aviar , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Melanoma Experimental/metabolismo , Ratones , Ratones Desnudos , Oncogenes , Plectina/genética , Sarcoma Aviar/genética
5.
FASEB J ; 36(2): e22153, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34997955

RESUMEN

DNA methylation is an epigenetic modification critical for the regulation of chromatin structure and gene expression during development and disease. The ten-eleven translocation (TET) enzyme family catalyzes the hydroxymethylation and subsequent demethylation of DNA by oxidizing 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC). Little is known about TET protein function due to a lack of pharmacological tools to manipulate DNA hydroxymethylation levels. In this study, we examined the role of TET-mediated DNA hydroxymethylation during BMP-induced C2C12 osteoblast differentiation using a novel cytosine-based selective TET enzyme inhibitor, Bobcat339 (BC339). Treatment of C2C12 cells with BC339 increased global 5mC and decreased global 5hmC without adversely affecting cell viability, proliferation, or apoptosis. Furthermore, BC339 treatment inhibited osteoblast marker gene expression and decreased alkaline phosphatase activity during differentiation. Methylated DNA immunoprecipitation and bisulfite sequencing showed that inhibition of TET with BC339 led to increased 5mC at specific CpG-rich regions at the promoter of Sp7, a key osteoblast transcription factor. Consistent with promoter 5mC marks being associated with transcriptional repression, luciferase activity of an Sp7-promoter-reporter construct was repressed by in vitro DNA methylation or BC339. Chromatin immunoprecipitation analysis confirmed that TET2 does indeed occupy the promoter region of Sp7. Accordingly, forced overexpression of SP7 rescued the inhibition of osteogenic differentiation by BC339. In conclusion, our data suggest that TET-mediated DNA demethylation of genomic regions, including the Sp7 promoter, plays a role in the initiation of osteoblast differentiation. Furthermore, BC339 is a novel pharmacological tool for the modulation of DNA methylation dynamics for research and therapeutic applications.


Asunto(s)
Diferenciación Celular/fisiología , ADN/metabolismo , Osteoblastos/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Células 3T3 , Animales , Apoptosis/fisiología , Biomarcadores/metabolismo , Línea Celular , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Desmetilación del ADN , Metilación de ADN/fisiología , Regulación de la Expresión Génica/fisiología , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoblastos/metabolismo , Regiones Promotoras Genéticas/genética
6.
J Biol Chem ; 296: 100790, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34019873

RESUMEN

Deletion of c-Src, a ubiquitously expressed tyrosine kinase, results in osteoclast dysfunction and osteopetrosis, in which bones harden into "stone." In contrast, deletion of the genes encoding other members of the Src family kinase (SFK) fails to produce an osteopetrotic phenotype. This suggests that c-Src performs a unique function in the osteoclast that cannot be compensated for by other SFKs. We aimed to identify the molecular basis of this unique role in osteoclasts and bone resorption. We found that c-Src, Lyn, and Fyn were the most highly expressed SFKs in WT osteoclasts, whereas Hck, Lck, Blk, and Fgr displayed low levels of expression. Formation of the podosome belt, clusters of unique actin assemblies, was disrupted in src-/- osteoclasts; introduction of constitutively activated SFKs revealed that only c-Src and Fyn could restore this process. To identify the key structural domains responsible, we constructed chimeric Src-Hck and Src-Lyn constructs in which the unique, SH3, SH2, or catalytic domains had been swapped. We found that the Src unique, SH3, and kinase domains were each crucial to establish Src functionality. The SH2 domain could however be substituted with Lyn or Hck SH2 domains. Furthermore, we demonstrate that c-Src's functionality is, in part, derived from an SH3-proximal proline-rich domain interaction with c-Cbl, leading to phosphorylation of c-Cbl Tyr700. These data help clarify Src's unique functionality in the organization of the cytoskeleton in osteoclasts, required for efficient bone resorption and explain why c-Src cannot be replaced, in osteoclasts, by other SFKs.


Asunto(s)
Osteoclastos/metabolismo , Podosomas/metabolismo , Dominios Homologos src , Familia-src Quinasas/metabolismo , Animales , Resorción Ósea/genética , Resorción Ósea/metabolismo , Diferenciación Celular , Células HEK293 , Humanos , Ratones , Osteoclastos/citología , Familia-src Quinasas/genética
7.
Sci Rep ; 11(1): 8418, 2021 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-33875709

RESUMEN

We have previously shown that parathyroid hormone (PTH) induces the phosphorylation of the DNA-binding protein Nascent polypeptide associated complex And Coregulator alpha (NACA), leading to nuclear translocation of NACA and activation of target genes. Using ChIP-Seq against NACA in parallel with RNA-sequencing, we report the identification of Ubiquitin Specific Peptidase 53 (Usp53) as a target gene of PTH-activated NACA in osteoblasts. A binding site for NACA within the ChIP fragment from the Usp53 promoter was confirmed by electrophoretic mobility shift assay. Activity of the Usp53 promoter (- 2325/+ 238 bp) was regulated by the JUN-CREB complex and this activation relied on activated PKA and the presence of NACA. Usp53 knockdown in ST2 stromal cells stimulated expression of the osteoblastic markers Bglap2 (Osteocalcin) and Alpl (Alkaline phosphatase) and inhibited expression of the adipogenic markers Pparg and Cebpa. A similar effect was measured when knocking down Naca. During osteoblastogenesis, the impact of Usp53 knockdown on PTH responses varied depending on the maturation stage of the cells. In vivo implantation of Usp53-knockdown bone marrow stromal cells in immunocompromised mice showed an increase in osteoblast number and a decrease in adipocyte counts. Our data suggest that Usp53 modulates the fate of mesenchymal cells by impacting lineage selection.


Asunto(s)
Adipocitos/metabolismo , Diferenciación Celular , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Adipogénesis , Fosfatasa Alcalina/metabolismo , Animales , Ratones , Osteocalcina/metabolismo , Hormona Paratiroidea/metabolismo
8.
Bone ; 132: 115209, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31866495

RESUMEN

Osteoclasts are multinuclear cells which maintain bone homeostasis by resorbing bone. During bone resorption, osteoclasts attach to the bone matrix via a sealing zone formed by an actin ring. Rous sarcoma oncogene (Src) is essential for actin ring formation and bone resorption. Recently, we demonstrated that plectin, a cytolinker protein, is a Src-binding protein in osteoclasts. However, the function of plectin in osteoclasts remains unknown. In this study, we demonstrated that shRNA knockdown of plectin in RAW 264.7 cells resulted in tartrate resistant acid phosphatase positive multinuclear cells (TRAP (+) MNCs) with impaired actin ring formation and bone resorption activity. Moreover, we found that in plectin-silenced TRAP (+) MNCs, Src and protein tyrosine kinase 2 beta (Pyk2), two critical kinases in osteoclastic bone resorption, were inactivated and microtubule polarity was disturbed. These results suggest that plectin plays a critical role in osteoclast biology by acting as a scaffold to facilitate Src and Pyk2 activation during microtubule organization.


Asunto(s)
Resorción Ósea , Quinasa 2 de Adhesión Focal , Células Cultivadas , Humanos , Microtúbulos , Osteoclastos , Plectina/genética
9.
J Biol Chem ; 294(20): 8184-8196, 2019 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-30948508

RESUMEN

The transcriptional cofactor nascent polypeptide-associated complex and co-regulator α (NACA) regulates osteoblast maturation and activity. NACA functions, at least in part, by binding to Jun proto-oncogene, AP-1 transcription factor subunit (cJUN) and potentiating the transactivation of AP-1 targets such as osteocalcin (Bglap) and matrix metallopeptidase 9 (Mmp9). NACA activity is modulated by phosphorylation carried out by several kinases, but a phosphatase regulating NACA's activity remains to be identified. Here, we used affinity purification with MS in HEK293T cells to isolate NACA complexes and identified protein phosphatase 1 catalytic subunit α (PP1A) as a NACA-associated Ser/Thr phosphatase. NACA interacted with multiple components of the PP1A holoenzyme complex: the PPP1CA catalytic subunit and the regulatory subunits PPP1R9B, PPP1R12A and PPP1R18. MS analysis revealed that NACA co-expression with PPP1CA causes dephosphorylation of NACA at Thr-89, Ser-151, and Thr-174. NACA Ser/Thr-to-alanine variants displayed increased nuclear localization, and NACA dephosphorylation was associated with specific recruitment of novel NACA interactants, such as basic transcription factor 3 (BTF3) and its homolog BTF3L4. NACA and PP1A cooperatively potentiated cJUN transcriptional activity of the AP-1-responsive MMP9-luciferase reporter, which was abolished when Thr-89, Ser-151, or Thr-174 were substituted with phosphomimetic aspartate residues. We confirmed the NACA-PP1A interaction in MC3T3-E1 osteoblastic cells and observed that NACA phosphorylation status at PP1A-sensitive sites is important for the regulation of AP-1 pathway genes and for osteogenic differentiation and matrix mineralization. These results suggest that PP1A dephosphorylates NACA at specific residues, impacting cJUN transcriptional activity and osteoblast differentiation and function.


Asunto(s)
Diferenciación Celular , Núcleo Celular/metabolismo , Chaperonas Moleculares/metabolismo , Osteoblastos/metabolismo , Proteína Fosfatasa 1/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transcripción Genética , Transporte Activo de Núcleo Celular/genética , Animales , Núcleo Celular/genética , Células HEK293 , Humanos , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Chaperonas Moleculares/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Osteoblastos/citología , Fosforilación/genética , Proteína Fosfatasa 1/genética , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-jun/genética , Elementos de Respuesta , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
Oncotarget ; 10(3): 404-414, 2019 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-30719233

RESUMEN

Melanoma, one of the most aggressive neoplasms, is characterized by rapid cell proliferation. Transducin-like Enhancer of Split (TLE) is an important regulator of cell proliferation via Histone deacetylase (HDAC) recruitment. Given that HDAC activity is associated with melanoma progression, we examined the relationship between TLE3, a TLE family member, and melanoma. TLE3 expression was increased during the progression of human patient melanoma (p < 0.05). Overexpression of Tle3 in B16 murine melanoma cells led to an increase in cell proliferation (p < 0.01) as well as the number of cyclinD1-positive cells. in vivo injection of mice with B16 cells overexpressing Tle3 resulted in larger tumor formation than in mice injected with control cells (p < 0.05). In contrast, siRNA-mediated knockdown of Tle3 in B16 cells or TLE3 in HMV-II human melanoma cells decreased proliferation (p < 0.01). Treatment of B16 cells with trichostatin A (2.5 µM), a class I and II HDAC inhibitor, prevented the effect s of Tle3 on proliferation. In conclusion, these data indicate that Tle3 is required, at least in part, for proliferation in the B16 mouse melanoma model.

11.
Mol Cell Biol ; 39(8)2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30692273

RESUMEN

Satellite cells (SCs) are skeletal muscle stem cells that proliferate in response to injury and provide myogenic precursors for growth and repair. Zfp423 is a transcriptional cofactor expressed in multiple immature cell populations, such as neuronal precursors, mesenchymal stem cells, and preadipocytes, where it regulates lineage allocation, proliferation, and differentiation. Here, we show that Zfp423 regulates myogenic progression during muscle regeneration. Zfp423 is undetectable in quiescent SCs but becomes expressed during SC activation. After expansion, Zfp423 is gradually downregulated as committed SCs terminally differentiate. Mice with satellite-cell-specific Zfp423 deletion exhibit severely impaired muscle regeneration following injury, with aberrant SC expansion, defective cell cycle exit, and failure to transition efficiently from the proliferative stage toward commitment. Consistent with a cell-autonomous role of Zfp423, shRNA-mediated knockdown of Zfp423 in myoblasts inhibits differentiation. Surprisingly, forced expression of Zfp423 in myoblasts induces differentiation into adipocytes and arrests myogenesis. Affinity purification of Zfp423 in myoblasts identified Satb2 as a nuclear partner of Zfp423 that cooperatively enhances Zfp423 transcriptional activity, which in turn affects myoblast differentiation. In conclusion, by controlling SC expansion and proliferation, Zfp423 is essential for muscle regeneration. Tight regulation of Zfp423 expression is essential for normal progression of muscle progenitors from proliferation to differentiation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/citología , Factores de Transcripción/metabolismo , Adipocitos/citología , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Proteínas de Unión al ADN/genética , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Desarrollo de Músculos/fisiología , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Regeneración/fisiología , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/fisiología , Transducción de Señal , Células Madre/citología , Factores de Transcripción/genética , Cicatrización de Heridas
12.
In Vivo ; 32(6): 1339-1344, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30348686

RESUMEN

BACKGROUND: The global incidence of diabetes mellitus (DM) has risen precipitously, even in middle- and low-income countries. Peroxisome proliferator-activated receptor γ (PPARγ) plays an important role in the control of cellular glucose metabolism. Activation of PPARγ beneficially results in increased insulin sensitivity. However, the expression of PPARγ is reduced by obesity and several nutritional factors. Here we examined the effect of geranylgeraniol (GGOH), a bioactive compound found naturally in fruits, vegetables, and grains, on the expression and activation of PPARγ. MATERIALS AND METHODS: C3H10T1/2 mouse embryonic fibroblasts and 3T3-L1 pre-adipocytes were used as in vitro models of adipocyte differentiation and function. Quantitative reverse-transcriptase polymerase chain reaction, western blotting, Oil Red O staining, and luciferase assay were performed to respectively assess mRNA expression, protein levels, lipid droplet formation and transcriptional activity. RESULTS: GGOH increased the expression of PPARγ in adipocyte lineage cells. GGOH also enhanced adipogenesis induced by rosiglitazone, a thiazolidinedione class PPARγ agonist. CONCLUSION: GGOH induces PPARγ expression and enhances the biological effects of a PPARγ agonist in adipocyte lineage cells.


Asunto(s)
Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Diterpenos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , PPAR gamma/agonistas , PPAR gamma/genética , Células 3T3-L1 , Animales , Fibroblastos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Ratones , PPAR gamma/metabolismo
13.
In Vivo ; 32(6): 1427-1431, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30348697

RESUMEN

BACKGROUND: Geranylgeraniol (GGOH) is a C20 isoprenoid found in fruits, vegetables, and grains, including rice. As a food substance, GGOH is categorized as 'Generally Recognized as Safe'. GGOH is an intermediate product in the mevalonate pathway and acts as a precursor to geranylgeranyl pyrophosphate. MATERIALS AND METHODS: C2C12 mouse myoblasts derived from muscle satellite cells were used. Quantitative reverse-transcriptase polymerase chain reaction, western blotting analysis, and immunocytochemical analysis were performed to respectively assess mRNA expression, protein levels, and the number of myofibers. RESULTS: GGOH reduced the expression levels of skeletal muscle atrophy-related ubiquitin ligases in myofibers derived from C2C12 cells. GGOH induced myogenic differentiation of C2C12 cells via geranylgeranylation. GGOH did not adversely affect the proliferation of C2C12 cells. CONCLUSION: GGOH induces myoblast differentiation in C2C12 cells.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Diterpenos/farmacología , Mioblastos/citología , Mioblastos/efectos de los fármacos , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Inmunohistoquímica , Redes y Vías Metabólicas/efectos de los fármacos , Ratones , Mioblastos/metabolismo
14.
J Biol Chem ; 292(31): 12885-12894, 2017 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-28607151

RESUMEN

Satellite cells are skeletal muscle stem cells that provide myonuclei for postnatal muscle growth, maintenance, and repair/regeneration in adults. Normally, satellite cells are mitotically quiescent, but they are activated in response to muscle injury, in which case they proliferate extensively and exhibit up-regulated expression of the transcription factor MyoD, a master regulator of myogenesis. MyoD forms a heterodimer with E proteins through their basic helix-loop-helix domain, binds to E boxes in the genome and thereby activates transcription at muscle-specific promoters. The central role of MyoD in muscle differentiation has increased interest in finding potential MyoD regulators. Here we identified transducin-like enhancer of split (TLE3), one of the Groucho/TLE family members, as a regulator of MyoD function during myogenesis. TLE3 was expressed in activated and proliferative satellite cells in which increased TLE3 levels suppressed myogenic differentiation, and, conversely, reduced TLE3 levels promoted myogenesis with a concomitant increase in proliferation. We found that, via its glutamine- and serine/proline-rich domains, TLE3 interferes with MyoD function by disrupting the association between the basic helix-loop-helix domain of MyoD and E proteins. Our findings indicate that TLE3 participates in skeletal muscle homeostasis by dampening satellite cell differentiation via repression of MyoD transcriptional activity.


Asunto(s)
Proteínas Co-Represoras/metabolismo , Regulación del Desarrollo de la Expresión Génica , Desarrollo de Músculos , Fibras Musculares Esqueléticas/metabolismo , Proteína MioD/antagonistas & inhibidores , Mioblastos/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Factor de Transcripción Activador 3/química , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Proteínas Co-Represoras/antagonistas & inhibidores , Proteínas Co-Represoras/química , Proteínas Co-Represoras/genética , Eliminación de Gen , Secuencias Hélice-Asa-Hélice , Masculino , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/citología , Proteína MioD/química , Proteína MioD/genética , Proteína MioD/metabolismo , Mioblastos/citología , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Interferencia de ARN , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Células Satélite del Músculo Esquelético/citología
15.
Mol Cell Biol ; 34(16): 3076-85, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24891617

RESUMEN

Osteoblasts and adipocytes arise from a common mesenchymal precursor cell. The cell fate decision of a mesenchymal precursor cell is under the influence of molecular cues and signaling pathways that lead to the activation or repression of lineage-specific transcription factors. The molecular mechanisms determining osteoblast versus adipocyte lineage specificity in response to bone morphogenic protein (BMP) remain unclear. In this study, we describe the mechanism through which Zfp521 (ZNF521), a regulator of lineage progression in multiple immature cell populations, regulates lineage specification of mesenchymal progenitor cells during BMP-induced differentiation events. In vivo deletion or in vitro knockdown of Zfp521 in mesenchymal precursors resulted in increased expression of the adipocyte determinant factor Zfp423 (ZNF423). This was concurrent with the loss of histone H3K9 methylation and an increase in histone H3K9 acetylation at the Zfp423 promoter, which together are indicative of decreased gene repression. Indeed, we found that Zfp521 occupies and represses the promoter and intronic enhancer regions of Zfp423. Accordingly, conditional deletion of Zfp521 inhibited heterotopic bone formation in response to local injection of BMP2. In contrast, marrow adiposity within BMP2-induced bone was markedly enhanced in Zfp521-deficient mice, suggesting that precursor cells lacking Zfp521 differentiate preferentially into adipocytes instead of osteoblasts in response to BMP2. Consistent with a cell-autonomous role of Zfp521 in mesenchymal precursors, knockdown of Zfp521 in stromal cells prevented BMP2-induced osteoblast marker expression and simultaneously enhanced lipid accumulation and expression of adipocyte-related genes. Taken together, the data suggest that Zfp521 is a cell fate switch critical for BMP-induced osteoblast commitment and identify Zfp521 as the intrinsic repressor of Zfp423 and hence of adipocyte commitment during BMP-induced mesenchymal precursor differentiation.


Asunto(s)
Adipocitos/metabolismo , Proteína Morfogenética Ósea 2/farmacología , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Osteoblastos/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Acetilación , Adipocitos/citología , Animales , Diferenciación Celular/genética , Línea Celular , Linaje de la Célula/genética , Metilación de ADN , Elementos de Facilitación Genéticos/genética , Células HEK293 , Histonas/genética , Humanos , Lípidos/biosíntesis , Células Madre Mesenquimatosas , Ratones , Ratones Noqueados , Osteoblastos/citología , Osteogénesis/genética , PPAR gamma/genética , Regiones Promotoras Genéticas/genética , Interferencia de ARN , Eliminación de Secuencia/genética
16.
Periodontol 2000 ; 63(1): 102-22, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23931057

RESUMEN

As broadly demonstrated for the formation of a functional skeleton, proper mineralization of periodontal alveolar bone and teeth - where calcium phosphate crystals are deposited and grow within an extracellular matrix - is essential for dental function. Mineralization defects in tooth dentin and cementum of the periodontium invariably lead to a weak (soft or brittle) dentition in which teeth become loose and prone to infection and are lost prematurely. Mineralization of the extremities of periodontal ligament fibers (Sharpey's fibers) where they insert into tooth cementum and alveolar bone is also essential for the function of the tooth-suspensory apparatus in occlusion and mastication. Molecular determinants of mineralization in these tissues include mineral ion concentrations (phosphate and calcium), pyrophosphate, small integrin-binding ligand N-linked glycoproteins and matrix vesicles. Amongst the enzymes important in regulating these mineralization determinants, two are discussed at length here, with clinical examples given, namely tissue-nonspecific alkaline phosphatase and phosphate-regulating gene with homologies to endopeptidases on the X chromosome. Inactivating mutations in these enzymes in humans and in mouse models lead to the soft bones and teeth characteristic of hypophosphatasia and X-linked hypophosphatemia, respectively, where the levels of local and systemic circulating mineralization determinants are perturbed. In X-linked hypophosphatemia, in addition to renal phosphate wasting causing low circulating phosphate levels, phosphorylated mineralization-regulating small integrin-binding ligand N-linked glycoproteins, such as matrix extracellular phosphoglycoprotein and osteopontin, and the phosphorylated peptides proteolytically released from them, such as the acidic serine- and aspartate-rich-motif peptide, may accumulate locally to impair mineralization in this disease.


Asunto(s)
Proceso Alveolar/fisiología , Calcificación Fisiológica/fisiología , Proteínas del Esmalte Dental/fisiología , Matriz Extracelular/fisiología , Raquitismo Hipofosfatémico Familiar/fisiopatología , Hipofosfatasia/fisiopatología , Ligamento Periodontal/fisiología , Fosfatasa Alcalina/fisiología , Proceso Alveolar/enzimología , Animales , Fosfatos de Calcio/metabolismo , Difosfatos/metabolismo , Modelos Animales de Enfermedad , Endopeptidasas/fisiología , Matriz Extracelular/enzimología , Humanos , Ligamento Periodontal/enzimología
17.
J Bone Miner Res ; 28(3): 688-99, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22991293

RESUMEN

X-linked hypophosphatemia (XLH/HYP)-with renal phosphate wasting, hypophosphatemia, osteomalacia, and tooth abscesses-is caused by mutations in the zinc-metallopeptidase PHEX gene (phosphate-regulating gene with homologies to endopeptidase on the X chromosome). PHEX is highly expressed by mineralized tissue cells. Inactivating mutations in PHEX lead to distal renal effects (implying accumulation of a secreted, circulating phosphaturic factor) and accumulation in bone and teeth of mineralization-inhibiting, acidic serine- and aspartate-rich motif (ASARM)-containing peptides, which are proteolytically derived from the mineral-binding matrix proteins of the SIBLING family (small, integrin-binding ligand N-linked glycoproteins). Although the latter observation suggests a local, direct matrix effect for PHEX, its physiologically relevant substrate protein(s) have not been identified. Here, we investigated two SIBLING proteins containing the ASARM motif-osteopontin (OPN) and bone sialoprotein (BSP)-as potential substrates for PHEX. Using cleavage assays, gel electrophoresis, and mass spectrometry, we report that OPN is a full-length protein substrate for PHEX. Degradation of OPN was essentially complete, including hydrolysis of the ASARM motif, resulting in only very small residual fragments. Western blotting of Hyp (the murine homolog of human XLH) mouse bone extracts having no PHEX activity clearly showed accumulation of an ∼35 kDa OPN fragment that was not present in wild-type mouse bone. Immunohistochemistry and immunogold labeling (electron microscopy) for OPN in Hyp bone likewise showed an accumulation of OPN and/or its fragments compared with normal wild-type bone. Incubation of Hyp mouse bone extracts with PHEX resulted in the complete degradation of these fragments. In conclusion, these results identify full-length OPN and its fragments as novel, physiologically relevant substrates for PHEX, suggesting that accumulation of mineralization-inhibiting OPN fragments may contribute to the mineralization defect seen in the osteomalacic bone characteristic of XLH/HYP.


Asunto(s)
Huesos/metabolismo , Raquitismo Hipofosfatémico Familiar/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X , Osteopontina/metabolismo , Endopeptidasa Neutra Reguladora de Fosfato PHEX/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting , Modelos Animales de Enfermedad , Electroforesis en Gel de Poliacrilamida , Inmunohistoquímica , Espectrometría de Masas , Ratones , Datos de Secuencia Molecular , Osteopontina/química , Proteolisis
18.
Biomaterials ; 31(36): 9422-30, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20943264

RESUMEN

Apatite-binding peptides discovered by phage display provide an alternative design method for creating functional biomaterials for bone and tooth tissue repair. A limitation of this approach is the absence of display peptide phosphorylation--a post-translational modification important to mineral-binding proteins. To refine the material specificity of a recently identified apatite-binding peptide, and to determine critical design parameters (net charge, charge distribution, amino acid sequence and composition) controlling peptide affinity for mineral, we investigated the effects of phosphorylation and sequence scrambling on peptide adsorption to four different apatites (bone-like mineral, and three types of apatite containing initially 0, 5.6 and 10.5% carbonate). Phosphorylation of the VTKHLNQISQSY peptide (VTK peptide) led to a 10-fold increase in peptide adsorption (compared to nonphosphorylated peptide) to bone-like mineral, and a 2-fold increase in adsorption to the carbonated apatite, but there was no effect of phosphorylation on peptide affinity to pure hydroxyapatite (without carbonate). Sequence scrambling of the nonphosphorylated VTK peptide enhanced its specificity for the bone-like mineral, but scrambled phosphorylated VTK peptide (pVTK) did not significantly alter mineral-binding suggesting that despite the importance of sequence order and/or charge distribution to mineral-binding, the enhanced binding after phosphorylation exceeds any further enhancement by altered sequence order. Osteoblast culture mineralization was dose-dependently inhibited by pVTK and to a significantly lesser extent by scrambled pVTK, while the nonphosphorylated and scrambled forms had no effect, indicating that inhibition of osteoblast mineralization is dependent on both peptide sequence and charge. Computational modeling of peptide-mineral interactions indicated a favorable change in binding energy upon phosphorylation that was unaffected by scrambling. In conclusion, phosphorylation of serine residues increases peptide specificity for bone-like mineral, whose adsorption is determined primarily by sequence composition and net charge as opposed to sequence order. However, sequence order in addition to net charge modulates the mineralization of osteoblast cultures. The ability of such peptides to inhibit mineralization has potential utility in the management of pathologic calcification.


Asunto(s)
Minerales/metabolismo , Péptidos/química , Péptidos/metabolismo , Fosfopéptidos/química , Fosfopéptidos/metabolismo , Adsorción , Secuencia de Aminoácidos , Animales , Calcificación Fisiológica , Diferenciación Celular , Línea Celular , Células Cultivadas , Colorantes Fluorescentes/metabolismo , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Osteoblastos/citología , Osteoblastos/metabolismo , Fosforilación
20.
Bone ; 46(4): 1100-7, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20079473

RESUMEN

Inositol hexakisphosphate (IP6, phytic acid) is an endogenous compound present in mammalian cells and tissues. Differentially phosphorylated forms of inositol are well-documented to have important roles in signal transduction, cell proliferation and differentiation, and IP6 in particular has been suggested to inhibit soft tissue calcification (specifically renal and vascular calcification) by binding extracellularly to calcium oxalate and calcium phosphate crystals. However, the effects of IP6 on bone mineralization are largely unknown. In this study, we used MC3T3-E1 osteoblast cultures to examine the effects of exogenous IP6 on osteoblast function and matrix mineralization. IP6 at physiologic concentrations caused a dose-dependent inhibition of mineralization without affecting cell viability, proliferation or collagen deposition. Osteoblast differentiation markers, including tissue-nonspecific alkaline phosphatase activity, bone sialoprotein and osteocalcin mRNA levels, were not adversely affected by IP6 treatment. On the other hand, IP6 markedly increased protein and mRNA levels of osteopontin, a potent inhibitor of crystal growth and matrix mineralization. Inositol alone (without phosphate), as well as inositol hexakis-sulphate, a compound with a high negative charge similar to IP6, had no effect on mineralization or osteopontin induction. Binding of IP6 to mineral crystals from the osteoblast cultures, as well as to synthetic hydroxyapatite crystals, was confirmed by a colorimetric assay for IP6. In summary, IP6 inhibits mineralization of osteoblast cultures by binding to growing crystals through negatively charged phosphate groups and by induction of inhibitory osteopontin expression. These data suggest that IP6 may regulate physiologic bone mineralization by directly acting extracellularly, and by serving as a specific signal at the cellular level for the regulation of osteopontin gene expression.


Asunto(s)
Calcificación Fisiológica/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Ácido Fítico/farmacología , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Western Blotting , Línea Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Sialoproteína de Unión a Integrina , Ratones , Osteoblastos/metabolismo , Osteocalcina/genética , Osteocalcina/metabolismo , Osteopontina/genética , Osteopontina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sialoglicoproteínas/genética , Sialoglicoproteínas/metabolismo , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...