Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
J Biol Chem ; 293(49): 19001-19011, 2018 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-30327434

RESUMEN

Ezh2 is a histone methyltransferase that suppresses osteoblast maturation and skeletal development. We evaluated the role of Ezh2 in chondrocyte lineage differentiation and endochondral ossification. Ezh2 was genetically inactivated in the mesenchymal, osteoblastic, and chondrocytic lineages in mice using the Prrx1-Cre, Osx1-Cre, and Col2a1-Cre drivers, respectively. WT and conditional knockout mice were phenotypically assessed by gross morphology, histology, and micro-CT imaging. Ezh2-deficient chondrocytes in micromass culture models were evaluated using RNA-Seq, histologic evaluation, and Western blotting. Aged mice with Ezh2 deficiency were also evaluated for premature development of osteoarthritis using radiographic analysis. Ezh2 deficiency in murine chondrocytes reduced bone density at 4 weeks of age but caused no other gross developmental effects. Knockdown of Ezh2 in chondrocyte micromass cultures resulted in a global reduction in trimethylation of histone 3 lysine 27 (H3K27me3) and altered differentiation in vitro RNA-Seq analysis revealed enrichment of an osteogenic gene expression profile in Ezh2-deficient chondrocytes. Joint development proceeded normally in the absence of Ezh2 in chondrocytes without inducing excessive hypertrophy or premature osteoarthritis in vivo In summary, loss of Ezh2 reduced H3K27me3 levels, increased the expression of osteogenic genes in chondrocytes, and resulted in a transient post-natal bone phenotype. Remarkably, Ezh2 activity is dispensable for normal chondrocyte maturation and endochondral ossification in vivo, even though it appears to have a critical role during early stages of mesenchymal lineage commitment.


Asunto(s)
Cartílago/metabolismo , Condrocitos/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/genética , Osteogénesis/fisiología , Animales , Diferenciación Celular/fisiología , Condrogénesis , Técnicas de Silenciamiento del Gen , Histonas/química , Histonas/metabolismo , Lisina/química , Metilación , Ratones , Transcriptoma
2.
J Biol Chem ; 293(33): 12894-12907, 2018 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-29899112

RESUMEN

Epigenetic mechanisms control skeletal development and osteoblast differentiation. Pharmacological inhibition of the histone 3 Lys-27 (H3K27) methyltransferase enhancer of zeste homolog 2 (EZH2) in WT mice enhances osteogenesis and stimulates bone formation. However, conditional genetic loss of Ezh2 early in the mesenchymal lineage (i.e. through excision via Prrx1 promoter-driven Cre) causes skeletal abnormalities due to patterning defects. Here, we addressed the key question of whether Ezh2 controls osteoblastogenesis at later developmental stages beyond patterning. We show that Ezh2 loss in committed pre-osteoblasts by Cre expression via the osterix/Sp7 promoter yields phenotypically normal mice. These Ezh2 conditional knock-out mice (Ezh2 cKO) have normal skull bones, clavicles, and long bones but exhibit increased bone marrow adiposity and reduced male body weight. Remarkably, in vivo Ezh2 loss results in a low trabecular bone phenotype in young mice as measured by micro-computed tomography and histomorphometry. Thus, Ezh2 affects bone formation stage-dependently. We further show that Ezh2 loss in bone marrow-derived mesenchymal cells suppresses osteogenic differentiation and impedes cell cycle progression as reflected by decreased metabolic activity, reduced cell numbers, and changes in cell cycle distribution and in expression of cell cycle markers. RNA-Seq analysis of Ezh2 cKO calvaria revealed that the cyclin-dependent kinase inhibitor Cdkn2a is the most prominent cell cycle target of Ezh2 Hence, genetic loss of Ezh2 in mouse pre-osteoblasts inhibits osteogenesis in part by inducing cell cycle changes. Our results suggest that Ezh2 serves a bifunctional role during bone formation by suppressing osteogenic lineage commitment while simultaneously facilitating proliferative expansion of osteoprogenitor cells.


Asunto(s)
Ciclo Celular/fisiología , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Osteoblastos/metabolismo , Osteogénesis/fisiología , Caracteres Sexuales , Animales , Proteína Potenciadora del Homólogo Zeste 2/genética , Femenino , Masculino , Ratones , Ratones Transgénicos , Osteoblastos/citología
3.
Am J Ind Med ; 62(4): 309-316, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30697792

RESUMEN

BACKGROUND: Minnesota has an ethnically diverse labor force, with the largest number of refugees per capita in the United States. In recent years, Minnesota has been and continues to be a major site for immigrant and refugee resettlement in the United States, with a large population of both immigrant and native born Hmong, Hispanic, and East Africans. This study seeks to evaluate the injury risk among the evolving minority workforce in the Minnesota Twin Cities region. METHODS: A retrospective cohort study identifying work-related injuries following pre-employment examinations was performed using electronic health records from a large multi-clinic occupational medicine practice. Preplacement examinations and subsequent work-related injuries were pulled from the electronic health record using representative ICD-10 codes for surveillance examinations and injuries. This study included patient records collected over a 2-year period from January 1, 2015, through December, 2016. The patients in this cohort worked in a wide-array of occupations including production, assembly, construction, law enforcement, among others. RESULTS: Hispanic minority workers were twice as likely to be injured at work compared with White workers. Hispanics were 2.89 times more likely to develop back injuries compared with non-Hispanic workers, and 1.86 times more likely to develop upper extremity injuries involving the hand, wrist, or elbow. CONCLUSION: Clinical practice data shows that Hispanic workers are at increased risk for work-related injuries in Minnesota. They were especially susceptible to back and upper extremity injuries. Lower injury rates in non-Hispanic minority workers, may be the result of injury underreporting and require further investigation.


Asunto(s)
Traumatismos del Brazo/etnología , Asiático/estadística & datos numéricos , Traumatismos de la Espalda/etnología , Negro o Afroamericano/estadística & datos numéricos , Traumatismos de la Mano/etnología , Hispánicos o Latinos/estadística & datos numéricos , Traumatismos Ocupacionales/etnología , Lesiones del Hombro/etnología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Minnesota/epidemiología , Grupos Minoritarios/estadística & datos numéricos , Medicina del Trabajo , Modelos de Riesgos Proporcionales , Estudios Retrospectivos , Población Blanca/estadística & datos numéricos , Adulto Joven
4.
J Cell Biochem ; 119(10): 8204-8219, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29923217

RESUMEN

Osteoblast differentiation is controlled by transcription factor RUNX2 which temporally activates or represses several bone-related genes, including those encoding extracellular matrix proteins or factors that control cell-cell, and cell-matrix interactions. Cell-cell communication in the many skeletal pericellular micro-niches is critical for bone development and involves paracrine secretion of growth factors and morphogens. This paracrine signaling is in part regulated by "A Disintegrin And Metalloproteinase" (ADAM) proteins. These cell membrane-associated metalloproteinases support proteolytic release ("shedding") of protein ectodomains residing at the cell surface. We analyzed microarray and RNA-sequencing data for Adam genes and show that Adam17, Adam10, and Adam9 are stimulated during BMP2 mediated induction of osteogenic differentiation and are robustly expressed in human osteoblastic cells. ADAM17, which was initially identified as a tumor necrosis factor alpha (TNFα) converting enzyme also called (TACE), regulates TNFα-signaling pathway, which inhibits osteoblast differentiation. We demonstrate that Adam17 expression is suppressed by RUNX2 during osteoblast differentiation through the proximal Adam17 promoter region (-0.4 kb) containing two functional RUNX2 binding motifs. Adam17 downregulation during osteoblast differentiation is paralleled by increased RUNX2 expression, cytoplasmic-nuclear translocation and enhanced binding to the Adam17 proximal promoter. Forced expression of Adam17 reduces Runx2 and Alpl expression, indicating that Adam17 may negatively modulate osteoblast differentiation. These findings suggest a novel regulatory mechanism involving a reciprocal Runx2-Adam17 negative feedback loop to regulate progression through osteoblast differentiation. Our results suggest that RUNX2 may control paracrine signaling through regulation of ectodomain shedding at the cell surface of osteoblasts by directly suppressing Adam17 expression.


Asunto(s)
Proteína ADAM17/genética , Proteína Morfogenética Ósea 2/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Retroalimentación Fisiológica , Osteoblastos/metabolismo , Osteogénesis/genética , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM10/genética , Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Sitios de Unión , Proteína Morfogenética Ósea 2/metabolismo , Diferenciación Celular , Línea Celular , Línea Celular Tumoral , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Osteoblastos/citología , Comunicación Paracrina/genética , Regiones Promotoras Genéticas , Unión Proteica , Ratas , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
5.
Connect Tissue Res ; 59(6): 542-549, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29513041

RESUMEN

PURPOSE: Revascularization of natural and synthetic scaffolds is a critical part of the scaffold's incorporation and tissue ingrowth. Our goals were to create a biocompatible polymer scaffold with 3D-printing technology, capable of sustaining vascularization and tissue ingrowth. METHODS: We synthesized biodegradable polycaprolactone fumarate (PCLF) scaffolds to allow tissue ingrowth via large interconnected pores. The scaffolds were prepared with Poly(lactic-co-glycolic acid)(PLGA) microspheres seeded with or without different growth factors including VEGF,FGF-2, and/or BMP-2. Scaffolds were implanted into the subcutaneous tissues of rats before undergoing histologic and microCT angiographic analysis. RESULTS: At harvest after 12 weeks, scaffolds had tissue infiltrating into their pores without signs of scar tissue formation, fibrous capsule formation, or immune responses against PCLF. Histology for M1/M2 macrophage phenotypes confirmed that there were no overt signs of immune responses. Both microCT angiography and histologic analysis demonstrated marked tissue and vessel ingrowth throughout the pores traversing the body of the scaffolds. Scaffolds seeded with microspheres containing VEGF or VEGF with either BMP-2 or FGF-2 had significantly higher vascular ingrowth and vessel penetration than controls. All VEGF-augmented scaffolds were positive for Factor-VIII and exhibited collagen tissue infiltration throughout the pores. Furthermore, scaffolds with VEGF and BMP-2 had high levels of mineral deposition throughout the scaffold that are attributable to BMP-2. CONCLUSIONS: PCLF polymer scaffold can be utilized as a framework for vascular ingrowth and regeneration of multiple types of tissues. This novel scaffold material has promise in tissue regeneration across all types of tissues from soft tissue to bone.


Asunto(s)
Neovascularización Fisiológica/efectos de los fármacos , Poliésteres , Impresión Tridimensional , Andamios del Tejido/química , Factor A de Crecimiento Endotelial Vascular , Animales , Proteína Morfogenética Ósea 2/química , Proteína Morfogenética Ósea 2/farmacología , Reactivos de Enlaces Cruzados/química , Fumaratos/química , Poliésteres/química , Poliésteres/farmacología , Ratas , Factor A de Crecimiento Endotelial Vascular/química , Factor A de Crecimiento Endotelial Vascular/farmacología
6.
J Biol Chem ; 291(47): 24594-24606, 2016 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-27758858

RESUMEN

Perturbations in skeletal development and bone degeneration may result in reduced bone mass and quality, leading to greater fracture risk. Bone loss is mitigated by bone protective therapies, but there is a clinical need for new bone-anabolic agents. Previous work has demonstrated that Ezh2 (enhancer of zeste homolog 2), a histone 3 lysine 27 (H3K27) methyltransferase, suppressed differentiation of osteogenic progenitors. Here, we investigated whether inhibition of Ezh2 can be leveraged for bone stimulatory applications. Pharmacologic inhibition and siRNA knockdown of Ezh2 enhanced osteogenic commitment of MC3T3 preosteoblasts. Next generation RNA sequencing of mRNAs and real time quantitative PCR profiling established that Ezh2 inactivation promotes expression of bone-related gene regulators and extracellular matrix proteins. Mechanistically, enhanced gene expression was linked to decreased H3K27 trimethylation (H3K27me3) near transcriptional start sites in genome-wide sequencing of chromatin immunoprecipitations assays. Administration of an Ezh2 inhibitor modestly increases bone density parameters of adult mice. Furthermore, Ezh2 inhibition also alleviated bone loss in an estrogen-deficient mammalian model for osteoporosis. Ezh2 inhibition enhanced expression of Wnt10b and Pth1r and increased the BMP-dependent phosphorylation of Smad1/5. Thus, these data suggest that inhibition of Ezh2 promotes paracrine signaling in osteoblasts and has bone-anabolic and osteoprotective potential in adults.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Osteoblastos/metabolismo , Osteogénesis , Osteoporosis/metabolismo , Comunicación Paracrina , Animales , Línea Celular , Proteína Potenciadora del Homólogo Zeste 2/genética , Femenino , Metilación/efectos de los fármacos , Ratones , Osteoblastos/patología , Osteoporosis/patología , Ovariectomía , ARN Interferente Pequeño/farmacología , Receptor de Hormona Paratiroídea Tipo 1 , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/genética , Proteína Smad5/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
7.
J Cell Biochem ; 118(11): 3662-3674, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28370561

RESUMEN

Osteosarcoma is the most common malignant bone tumor in children and adolescents. Metastasis and poor responsiveness to chemotherapy in osteosarcoma correlates with over-expression of the runt-related transcription factor RUNX2, which normally plays a key role in osteogenic lineage commitment, osteoblast differentiation, and bone formation. Furthermore, WNT/ß-catenin signaling is over-activated in osteosarcoma and promotes tumor progression. Importantly, the WNT/ß-catenin pathway normally activates RUNX2 gene expression during osteogenic lineage commitment. Therefore, we examined whether the WNT/ß-catenin pathway controls the tumor-related elevation of RUNX2 expression in osteosarcoma. We analyzed protein levels and nuclear localization of ß-catenin and RUNX2 in a panel of human osteosarcoma cell lines (SAOS, MG63, U2OS, HOS, G292, and 143B). In all six cell lines, ß-catenin and RUNX2 are expressed to different degrees and localized in the nucleus and/or cytoplasm. SAOS cells have the highest levels of RUNX2 protein that is localized in the nucleus, while MG63 cells have the lowest RUNX2 levels which is mostly localized in the cytoplasm. Levels of ß-catenin and RUNX2 protein are enhanced in HOS, G292, and 143B cells after treatment with the GSK3ß inhibitor SB216763. Furthermore, small interfering RNA (siRNA)-mediated depletion of ß-catenin inhibits RUNX2 expression in G292 cells. Thus, WNT/ß-catenin activation is required for RUNX2 expression in at least some osteosarcoma cell types, where RUNX2 is known to promote expression of metastasis related genes. J. Cell. Biochem. 118: 3662-3674, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Neoplasias Óseas/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/biosíntesis , Proteínas de Neoplasias/biosíntesis , Osteosarcoma/metabolismo , Vía de Señalización Wnt , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Línea Celular Tumoral , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Osteosarcoma/genética , Osteosarcoma/patología
8.
Biochem Biophys Res Commun ; 486(2): 307-313, 2017 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-28300555

RESUMEN

Osteosarcoma survival rate has not improved over the past three decades, and the debilitating side effects of the surgical treatment suggest the need for alternative local control approaches. Radiotherapy is largely ineffective in osteosarcoma, indicating a potential role for radiosensitizers. Blocking DNA repair, particularly by inhibiting the catalytic subunit of DNA-dependent protein kinase (DNA-PKCS), is an attractive option for the radiosensitization of osteosarcoma. In this study, the expression of DNA-PKCS in osteosarcoma tissue specimens and cell lines was examined. Moreover, the small molecule DNA-PKCS inhibitor, KU60648, was investigated as a radiosensitizing strategy for osteosarcoma cells in vitro. DNA-PKCS was consistently expressed in the osteosarcoma tissue specimens and cell lines studied. Additionally, KU60648 effectively sensitized two of those osteosarcoma cell lines (143B cells by 1.5-fold and U2OS cells by 2.5-fold). KU60648 co-treatment also altered cell cycle distribution and enhanced DNA damage. Cell accumulation at the G2/M transition point increased by 55% and 45%, while the percentage of cells with >20 γH2AX foci were enhanced by 59% and 107% for 143B and U2OS cells, respectively. These results indicate that the DNA-PKCS inhibitor, KU60648, is a promising radiosensitizing agent for osteosarcoma.


Asunto(s)
Neoplasias Óseas/terapia , Cromonas/farmacología , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica , Proteínas Nucleares/antagonistas & inhibidores , Osteosarcoma/terapia , Inhibidores de Proteínas Quinasas/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Neoplasias Óseas/enzimología , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Línea Celular Tumoral , Cromonas/química , Cromonas/metabolismo , Daño del ADN , Proteína Quinasa Activada por ADN/genética , Proteína Quinasa Activada por ADN/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de la radiación , Rayos gamma/uso terapéutico , Histonas/genética , Histonas/metabolismo , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Osteosarcoma/enzimología , Osteosarcoma/genética , Osteosarcoma/patología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/metabolismo , Fármacos Sensibilizantes a Radiaciones/química , Fármacos Sensibilizantes a Radiaciones/metabolismo , Análisis de Secuencia de ARN
9.
J Biol Chem ; 290(46): 27604-17, 2015 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-26424790

RESUMEN

Epigenetic control of gene expression is critical for normal fetal development. However, chromatin-related mechanisms that activate bone-specific programs during osteogenesis have remained underexplored. Therefore, we investigated the expression profiles of a large cohort of epigenetic regulators (>300) during osteogenic differentiation of human mesenchymal cells derived from the stromal vascular fraction of adipose tissue (AMSCs). Molecular analyses establish that the polycomb group protein EZH2 (enhancer of zeste homolog 2) is down-regulated during osteoblastic differentiation of AMSCs. Chemical inhibitor and siRNA knockdown studies show that EZH2, a histone methyltransferase that catalyzes trimethylation of histone 3 lysine 27 (H3K27me3), suppresses osteogenic differentiation. Blocking EZH2 activity promotes osteoblast differentiation and suppresses adipogenic differentiation of AMSCs. High throughput RNA sequence (mRNASeq) analysis reveals that EZH2 inhibition stimulates cell cycle inhibitory proteins and enhances the production of extracellular matrix proteins. Conditional genetic loss of Ezh2 in uncommitted mesenchymal cells (Prrx1-Cre) results in multiple defects in skeletal patterning and bone formation, including shortened forelimbs, craniosynostosis, and clinodactyly. Histological analysis and mRNASeq profiling suggest that these effects are attributable to growth plate abnormalities and premature cranial suture closure because of precocious maturation of osteoblasts. We conclude that the epigenetic activity of EZH2 is required for skeletal patterning and development, but EZH2 expression declines during terminal osteoblast differentiation and matrix production.


Asunto(s)
Epigénesis Genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Osteogénesis/genética , Complejo Represivo Polycomb 2/metabolismo , Tejido Adiposo/citología , Animales , Tipificación del Cuerpo/genética , Huesos/embriología , Diferenciación Celular/genética , Línea Celular , Proteína Potenciadora del Homólogo Zeste 2 , Placa de Crecimiento/anomalías , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Humanos , Células Madre Mesenquimatosas/citología , Ratones , Osteoblastos/citología , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Complejo Represivo Polycomb 2/genética , ARN Interferente Pequeño/genética
10.
FASEB J ; 29(4): 1344-59, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25491310

RESUMEN

Serum amyloid A (A-SAA/Saa3) was shown before to affect osteoblastic metabolism. Here, using RT-quantitative PCR and/or immunoblotting, we show that expression of mouse Saa3 and human SAA1 and SAA2 positively correlates with increased cellular maturation toward the osteocyte phenotype. Expression is not detected in C3H10T1/2 embryonic fibroblasts but is successively higher in preosteoblastic MC3T3-E1 cells, late osteoblastic MLO-A5 cells, and MLO-Y4 osteocytes, consistent with findings using primary bone cells from newborn mouse calvaria. Recombinant Saa3 protein functionally inhibits osteoblast differentiation as reflected by reductions in the expression of osteoblast markers and decreased mineralization in newborn mouse calvaria. Yet, Saa3 protein enhances osteoclastogenesis in mouse macrophages/monocytes based on the number of multinucleated and tartrate-resistant alkaline phosphatase-positive cells and Calcr mRNA expression. Depletion of Saa3 in MLO osteocytes results in the loss of the mature osteocyte phenotype. Recombinant osteocalcin, which is reciprocally regulated with Saa3 at the osteoblast/osteocyte transition, attenuates Saa3 expression in MLO-Y4 osteocytes. Mechanistically, Saa3 produced by MLO-Y4 osteocytes is integrated into the extracellular matrix of MC3T3-E1 osteoblasts, where it associates with the P2 purinergic receptor P2rx7 to stimulate Mmp13 expression via the P2rx7/MAPK/ERK/activator protein 1 axis. Our data suggest that Saa3 may function as an important coupling factor in bone development and homeostasis.


Asunto(s)
Huesos/metabolismo , Proteína Amiloide A Sérica/metabolismo , Células 3T3 , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Huesos/citología , Diferenciación Celular , Línea Celular , Células Cultivadas , Homeostasis , Humanos , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Osteoblastos/citología , Osteoblastos/metabolismo , Osteocalcina/genética , Osteocalcina/metabolismo , Osteoclastos/citología , Osteoclastos/metabolismo , Osteocitos/citología , Osteocitos/metabolismo , Osteogénesis , Comunicación Paracrina , Filogenia , ARN Interferente Pequeño/genética , Receptores Purinérgicos P2X7/metabolismo , Homología de Secuencia de Aminoácido , Proteína Amiloide A Sérica/antagonistas & inhibidores , Proteína Amiloide A Sérica/genética , Cráneo/citología , Cráneo/metabolismo
11.
J Cell Physiol ; 230(1): 52-62, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24912092

RESUMEN

Human adipose-derived mesenchymal stromal cells (AMSCs) grown in platelet lysate are promising agents for therapeutic tissue regeneration. Here, we investigated whether manipulation of epigenetic events by the clinically relevant histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA) alters differentiation of AMSCs. The multipotency of AMSCs was validated by their ability to differentiate into osteogenic, chondrogenic, and adipogenic lineages. High-throughput RNA sequencing and RT-qPCR established that human histone deacetylases (HDAC1 to HDAC11, and SIRT1 to SIRT7) are differentially expressed in AMSCs. SAHA induces hyper-acetylation of histone H3 and H4, stimulates protein expression of the HDAC-responsive gene SLC9A3R1/NHERF1 and modulates the AKT/FOXO1 pathway. Biologically, SAHA interferes with osteogenic, chondrogenic and adipogenic lineage commitment of multipotent AMSCs. Mechanistically, SAHA-induced loss of differentiation potential of uncommitted AMSCs correlates with multiple changes in the expression of principal transcription factors that control mesenchymal or pluripotent states. We propose that SAHA destabilizes the multi-potent epigenetic state of uncommitted human AMSCs by hyper-acetylation and perturbation of key transcription factor pathways. Furthermore, AMSCs grown in platelet lysate may provide a useful biological model for screening of new HDAC inhibitors that control the biological fate of human mesenchymal stromal cells.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/biosíntesis , Ácidos Hidroxámicos/farmacología , Células Madre Mesenquimatosas/citología , Acetilación , Adipocitos/citología , Tejido Adiposo/citología , Secuencia de Bases , Células Cultivadas , Condrocitos/citología , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Regeneración Tisular Dirigida , Secuenciación de Nucleótidos de Alto Rendimiento , Histonas/metabolismo , Humanos , Osteoblastos/citología , Fosfoproteínas/biosíntesis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Análisis de Secuencia de ARN , Intercambiadores de Sodio-Hidrógeno/biosíntesis , Vorinostat
12.
J Cell Physiol ; 229(12): 2049-56, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24812032

RESUMEN

Ewing sarcoma is an aggressive pediatric small round cell tumor that predominantly occurs in bone. Approximately 85% of Ewing sarcomas harbor the EWS/FLI fusion protein, which arises from a chromosomal translocation, t(11:22)(q24:q12). EWS/FLI interacts with numerous lineage-essential transcription factors to maintain mesenchymal progenitors in an undifferentiated state. We previously showed that EWS/FLI binds the osteogenic transcription factor RUNX2 and prevents osteoblast differentiation. In this study, we investigated the role of another Runt-domain protein, RUNX3, in Ewing sarcoma. RUNX3 participates in mesenchymal-derived bone formation and is a context dependent tumor suppressor and oncogene. RUNX3 was detected in all Ewing sarcoma cells examined, whereas RUNX2 was detected in only 73% of specimens. Like RUNX2, RUNX3 binds to EWS/FLI via its Runt domain. EWS/FLI prevented RUNX3 from activating the transcription of a RUNX-responsive reporter, p6OSE2. Stable suppression of RUNX3 expression in the Ewing sarcoma cell line A673 delayed colony growth in anchorage independent soft agar assays and reversed expression of EWS/FLI-responsive genes. These results demonstrate an important role for RUNX3 in Ewing sarcoma.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/biosíntesis , Neoplasias de Tejido Óseo/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína EWS de Unión a ARN/metabolismo , Sarcoma de Ewing/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias de Tejido Óseo/patología , Proteínas de Fusión Oncogénica/genética , Proteína Proto-Oncogénica c-fli-1/genética , Proteína EWS de Unión a ARN/genética , Sarcoma de Ewing/patología
13.
J Cell Biochem ; 115(10): 1816-28, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24905804

RESUMEN

Improving the effectiveness of adipose-tissue derived human mesenchymal stromal/stem cells (AMSCs) for skeletal therapies requires a detailed characterization of mechanisms supporting cell proliferation and multi-potency. We investigated the molecular phenotype of AMSCs that were either actively proliferating in platelet lysate or in a basal non-proliferative state. Flow cytometry combined with high-throughput RNA sequencing (RNASeq) and RT-qPCR analyses validate that AMSCs express classic mesenchymal cell surface markers (e.g., CD44, CD73/NT5E, CD90/THY1, and CD105/ENG). Expression of CD90 is selectively elevated at confluence. Self-renewing AMSCs express a standard cell cycle program that successively mediates DNA replication, chromatin packaging, cyto-architectural enlargement, and mitotic division. Confluent AMSCs preferentially express genes involved in extracellular matrix (ECM) formation and cellular communication. For example, cell cycle-related biomarkers (e.g., cyclins E2 and B2, transcription factor E2F1) and histone-related genes (e.g., H4, HINFP, NPAT) are elevated in proliferating AMSCs, while ECM genes are strongly upregulated (>10-fold) in quiescent AMSCs. AMSCs also express pluripotency genes (e.g., POU5F1, NANOG, KLF4) and early mesenchymal markers (e.g., NES, ACTA2) consistent with their multipotent phenotype. Strikingly, AMSCs modulate expression of WNT signaling components and switch production of WNT ligands (from WNT5A/WNT5B/WNT7B to WNT2/WNT2B), while upregulating WNT-related genes (WISP2, SFRP2, and SFRP4). Furthermore, post-proliferative AMSCs spontaneously express fibroblastic, osteogenic, chondrogenic, and adipogenic biomarkers when maintained in confluent cultures. Our findings validate the biological properties of self-renewing and multi-potent AMSCs by providing high-resolution quality control data that support their clinical versatility.


Asunto(s)
Tejido Adiposo/citología , Condrogénesis/genética , Células Madre Mesenquimatosas/citología , Osteogénesis/genética , Adipogénesis/genética , Secuencia de Bases , Comunicación Celular/genética , Puntos de Control del Ciclo Celular/genética , Diferenciación Celular , Proliferación Celular/genética , Tratamiento Basado en Trasplante de Células y Tejidos , Replicación del ADN/genética , Matriz Extracelular/genética , Citometría de Flujo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunofenotipificación , Factor 4 Similar a Kruppel , Proteínas de la Membrana/metabolismo , Mitosis/genética , Análisis de Secuencia de ARN , Antígenos Thy-1/biosíntesis
14.
BMC Cancer ; 12: 93, 2012 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-22429849

RESUMEN

BACKGROUND: Osteosarcoma is a bone tumor that often affects children and young adults. Although a combination of surgery and chemotherapy has improved the survival rate in the past decades, local recurrence and metastases still develop in 40% of patients. A definite therapy is yet to be determined for osteosarcoma. Anti- tumor compound and a metabolite of estrogen, 2-methoxyestradiol (2-ME) induces cell death in osteosarcoma cells. In this report, we have investigated whether interferon (IFN) pathway is involved in 2-ME-induced anti-tumor effects in osteosarcoma cells. METHODS: 2-ME effects on IFN mRNA levels were determined by Real time PCR analysis. Transient transfections followed by reporter assays were used for investigating 2-ME effects on IFN-pathway. Western blot analyses were used to measure protein and phosphorylation levels of IFN-regulated eukaryotic initiation factor-2 alpha (eIF-2α). RESULTS: 2-ME regulates IFN and IFN-mediated effects in osteosarcoma cells. 2 -ME induces IFN gene activity and expression in osteosarcoma cells. 2-ME treatment induced IFN-stimulated response element (ISRE) sequence-dependent transcription and gamma-activated sequence (GAS)-dependent transcription in several osteosarcoma cells. Whereas, 2-ME did not affect IFN gene and IFN pathways in normal primary human osteoblasts (HOB). 2-ME treatment increased the phosphorylation of eIF-2α in osteosarcoma cells. Furthermore, analysis of osteosarcoma tissues shows that the levels of phosphorylated form of eIF-2α are decreased in tumor compared to normal controls. CONCLUSIONS: 2-ME treatment triggers the induction and activity of IFN and IFN pathway genes in 2-ME-sensitive osteosarcoma tumor cells but not in 2-ME-resistant normal osteoblasts. In addition, IFN-signaling is inhibited in osteosarcoma patients. Thus, IFN pathways play a role in osteosarcoma and in 2-ME-mediated anti-proliferative effects, and therefore targeted induction of IFN signaling could lead to effective treatment strategies in the control of osteosarcoma.


Asunto(s)
Neoplasias Óseas/metabolismo , Estradiol/análogos & derivados , Interferones/metabolismo , Osteosarcoma/metabolismo , Transducción de Señal/efectos de los fármacos , 2-Metoxiestradiol , Neoplasias Óseas/genética , Línea Celular Tumoral , Estradiol/farmacología , Factor 2 Eucariótico de Iniciación/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Interferones/genética , Osteosarcoma/genética , Regiones Promotoras Genéticas/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
15.
Stem Cells Dev ; 30(11): 601-609, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33757300

RESUMEN

An efficient musculoskeletal system depends on the precise assembly and coordinated growth and function of muscles, skeleton, and tendons. However, the mechanisms that drive integrated musculoskeletal development and coordinated growth and differentiation of each of these tissues are still being uncovered. Epigenetic modifiers have emerged as critical regulators of cell fate differentiation, but so far almost nothing is known about their roles in tendon biology. Previous studies have shown that epigenetic modifications driven by Enhancer of zeste homolog 2 (EZH2), a major histone methyltransferase, have significant roles in vertebrate development including skeletal patterning and bone formation. We now find that targeting Ezh2 through the limb mesenchyme also has significant effects on tendon and muscle patterning, likely reflecting the essential roles of early mesenchymal cues mediated by Ezh2 for coordinated patterning and development of all tissues of the musculoskeletal system. Conversely, loss of Ezh2 in the tendon cells did not disrupt overall tendon structure or collagen organization suggesting that tendon differentiation and maturation are independent of Ezh2 signaling.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2 , Osteogénesis , Diferenciación Celular/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Mesodermo , Osteogénesis/genética , Tendones
16.
J Orthop Res ; 36(5): 1305-1312, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28921656

RESUMEN

Chronic low back pain is a major cause of disability and health care costs. Effective treatments are inadequate for many patients. Animal models are essential to further understanding of the pain mechanism and testing potential therapies. Currently, a number of preclinical models have been developed attempting to mimic aspects of clinical conditions that contribute to low back pain (LBP). This review focused on describing these animal models and the main behavioral tests for assessing pain in each model. Animal models of LBP can be divided into the following five categories: Discogenic LBP, radicular back pain, facet joint osteoarthritis back pain, muscle-induced LBP, and spontaneous occurring LBP models. These models are important not only for enhancing our knowledge of how LBP is generated, but also for the development of novel therapeutic regimens to treat LBP in patients. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1305-1312, 2018.


Asunto(s)
Modelos Animales de Enfermedad , Dolor de la Región Lumbar/etiología , Dolor de la Región Lumbar/terapia , Animales , Ganglios Espinales/fisiología , Humanos , Hiperalgesia/fisiopatología , Ratones Transgénicos , Osteoartritis/fisiopatología , Dimensión del Dolor
17.
Gene Rep ; 10: 149-156, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29868646

RESUMEN

PURPOSE: The poor healing potential of intra-articular ligament injuries drives a need for the development of novel, viable 'neo-ligament' alternatives. Ex vivo approaches combining stem cell engineering, 3-dimensional biocompatible scaffold design and enhancement of biological and biomechanical functionality via the introduction of key growth factors and morphogens, represent a promising solution to ligament regeneration. METHODS: We investigated growth, differentiation and extracellular matrix (ECM) protein production of human adipose-derived mesenchymal stem/stromal cells (MSCs), cultured in 5% human platelet lysate (PL) and seeded on three-dimensional polycaprolactone (PCL) scaffolds, in response to the connective-tissue related ligands fibroblast growth factor 2 (basic) (FGF2) and growth and differentiation factor-5 (GDF5). Phenotypic alterations of MSCs under different biological conditions were examined using cell viability assays, real time qPCR analysis of total RNA, as well as immunofluorescence microscopy. RESULTS: Phenotypic conversion of MSCs into ECM producing fibroblastic cells proceeds spontaneously in the presence of human platelet lysate. Administration of FGF2 and/or GDF5 enhances production of mRNAs for several ECM proteins including Collagen types I and III, as well as Tenomodulin (e.g., COL1A1, TNMD), but not Tenascin-C (TNC). Differences in the in situ deposition of ECM proteins Collagen type III and Tenascin-C were validated by immunofluorescence microscopy. SUMMARY: Treatment of MSCs with FGF2 and GDF5 was not synergistic and occasionally antagonistic for ECM production. Our results suggest that GDF5 alone enhances the conversion of MSCs to fibroblastic cells possessing a phenotype consistent with that of connective-tissue fibroblasts.

18.
Gene ; 668: 87-96, 2018 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-29775757

RESUMEN

The physis is a well-established and anatomically distinct cartilaginous structure that is crucial for normal long-bone development and growth. Abnormalities in physis function are linked to growth plate disorders and other pediatric musculoskeletal diseases. Understanding the molecular pathways operative in the physis may permit development of regenerative therapies to complement surgically-based procedures that are the current standard of care for growth plate disorders. Here, we performed next generation RNA sequencing on mRNA isolated from human physis and other skeletal tissues (e.g., articular cartilage and bone; n = 7 for each tissue). We observed statistically significant enrichment of gene sets in the physis when compared to the other musculoskeletal tissues. Further analysis of these upregulated genes identified physis-specific networks of extracellular matrix proteins including collagens (COL2A1, COL6A1, COL9A1, COL14A1, COL16A1) and matrilins (MATN1, MATN2, MATN3), and signaling proteins in the WNT pathway (WNT10B, FZD1, FZD10, DKK2) or the FGF pathway (FGF10, FGFR4). Our results provide further insight into the gene expression networks that contribute to the physis' unique structural composition and regulatory signaling networks. Physis-specific expression profiles may guide ongoing initiatives in tissue engineering and cell-based therapies for treatment of growth plate disorders and growth modulation therapies. Furthermore, our findings provide new leads for therapeutic drug discovery that would permit future intervention through pharmacological rather than surgical strategies.


Asunto(s)
Cartílago/metabolismo , Transcriptoma , Biomarcadores , Huesos/metabolismo , Cartílago Articular/metabolismo , Colágeno/metabolismo , Perfilación de la Expresión Génica , Músculos/metabolismo , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN , Transducción de Señal
19.
Nat Commun ; 9(1): 3634, 2018 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-30194383

RESUMEN

Tissue-specific gene expression defines cellular identity and function, but knowledge of early human development is limited, hampering application of cell-based therapies. Here we profiled 5 distinct cell types at a single fetal stage, as well as chondrocytes at 4 stages in vivo and 2 stages during in vitro differentiation. Network analysis delineated five tissue-specific gene modules; these modules and chromatin state analysis defined broad similarities in gene expression during cartilage specification and maturation in vitro and in vivo, including early expression and progressive silencing of muscle- and bone-specific genes. Finally, ontogenetic analysis of freshly isolated and pluripotent stem cell-derived articular chondrocytes identified that integrin alpha 4 defines 2 subsets of functionally and molecularly distinct chondrocytes characterized by their gene expression, osteochondral potential in vitro and proliferative signature in vivo. These analyses provide new insight into human musculoskeletal development and provide an essential comparative resource for disease modeling and regenerative medicine.


Asunto(s)
Condrocitos/metabolismo , Condrogénesis , Mioblastos/metabolismo , Osteoblastos/metabolismo , Tenocitos/metabolismo , Animales , Biomarcadores/metabolismo , Epigénesis Genética , Desarrollo Fetal , Perfilación de la Expresión Génica , Código de Histonas , Humanos , Ratones , Análisis de Secuencia de ARN , Porcinos , Transcripción Genética , Transcriptoma
20.
J Orthop Res ; 36(5): 1356-1369, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29227558

RESUMEN

Degenerative disk disease of the spine is a major cause of back pain and disability. Optimization of regenerative medical therapies for degenerative disk disease requires a deep mechanistic understanding of the factors controlling the structural integrity of spinal tissues. In this investigation, we sought to identify candidate regulatory genes controlling extracellular matrix synthesis in spinal tissues. To achieve this goal we performed high throughput next generation RNA sequencing on 39 annulus fibrosus and 21 nucleus pulposus human tissue samples. Specimens were collected from patients undergoing surgical discectomy for the treatment of degenerative disk disease. Our studies identified associations between extracellular matrix genes, growth factors, and other important regulatory molecules. The fibrous matrix characteristic of annulus fibrosus was associated with expression of the growth factors platelet derived growth factor beta (PDGFB), vascular endothelial growth factor C (VEGFC), and fibroblast growth factor 9 (FGF9). Additionally we observed high expression of multiple signaling proteins involved in the NOTCH and WNT signaling cascades. Nucleus pulposus extracellular matrix related genes were associated with the expression of numerous diffusible growth factors largely associated with the transforming growth signaling cascade, including transforming factor alpha (TGFA), inhibin alpha (INHA), inhibin beta A (INHBA), bone morphogenetic proteins (BMP2, BMP6), and others. CLINICAL SIGNIFICANCE: this investigation provides important data on extracellular matrix gene regulatory networks in disk tissues. This information can be used to optimize pharmacologic, stem cell, and tissue engineering strategies for regeneration of the intervertebral disk and the treatment of back pain. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1356-1369, 2018.


Asunto(s)
Matriz Extracelular/metabolismo , Redes Reguladoras de Genes , Disco Intervertebral/metabolismo , Análisis de Secuencia de ARN , Adulto , Anciano , Anillo Fibroso/metabolismo , Humanos , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA