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

Intervalo de año de publicación
1.
J Cell Physiol ; 234(8): 13659-13679, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30637720

RESUMEN

Osteosarcomas are bone tumors that frequently metastasize to the lung. Aberrant expression of the transcription factor, runt-related transcription factor 2 (RUNX2), is a key pathological feature in osteosarcoma and associated with loss of p53 and miR-34 expression. Elevated RUNX2 may transcriptionally activate genes mediating tumor progression and metastasis, including the RUNX2 target gene osteopontin (OPN/SPP1). This gene encodes a secreted matricellular protein produced by osteoblasts to regulate bone matrix remodeling and tissue calcification. Here we investigated whether and how the RUNX2/OPN axis regulates lung metastasis of osteosarcoma. Importantly, RUNX2 depletion attenuates lung metastasis of osteosarcoma cells in vivo. Using next-generation RNA-sequencing, protein-based assays, as well as the loss- and gain-of-function approaches in selected osteosarcoma cell lines, we show that osteopontin messenger RNA levels closely correlate with RUNX2 expression and that RUNX2 controls the levels of secreted osteopontin. Elevated osteopontin levels promote heterotypic cell-cell adhesion of osteosarcoma cells to human pulmonary microvascular endothelial cells, but not in the presence of neutralizing antibodies. Collectively, these findings indicate that the RUNX2/OPN axis regulates the ability of osteosarcoma cells to attach to pulmonary endothelial cells as a key step in metastasis of osteosarcoma cells to the lung.


Asunto(s)
Neoplasias Óseas/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Invasividad Neoplásica , Osteopontina/metabolismo , Osteosarcoma/metabolismo , Animales , Neoplasias Óseas/patología , Adhesión Celular/fisiología , Línea Celular Tumoral , Células Endoteliales/metabolismo , Xenoinjertos , Humanos , Pulmón/metabolismo , Pulmón/patología , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos NOD , Ratones SCID , Osteosarcoma/secundario
2.
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
3.
J Cell Biochem ; 118(5): 1262-1272, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27862226

RESUMEN

Osteogenic lineage commitment and progression is controlled by multiple signaling pathways (e.g., WNT, BMP, FGF) that converge on bone-related transcription factors. Access of osteogenic transcription factors to chromatin is controlled by epigenetic regulators that generate post-translational modifications of histones ("histone code"), as well as read, edit and/or erase these modifications. Our understanding of the biological role of epigenetic regulators in osteoblast differentiation remains limited. Therefore, we performed next-generation RNA sequencing (RNA-seq) and established which chromatin-related proteins are robustly expressed in mouse bone tissues (e.g., fracture callus, calvarial bone). These studies also revealed that cells with increased osteogenic potential have higher levels of the H4K20 methyl transferase Suv420h2 compared to other methyl transferases (e.g., Suv39h1, Suv39h2, Suv420h1, Ezh1, Ezh2). We find that all six epigenetic regulators are transiently expressed at different stages of osteoblast differentiation in culture, with maximal mRNAs levels of Suv39h1 and Suv39h2 (at day 3) preceding maximal expression of Suv420h1 and Suv420h2 (at day 7) and developmental stages that reflect, respectively, early and later collagen matrix deposition. Loss of function analysis of Suv420h2 by siRNA depletion shows loss of H4K20 methylation and decreased expression of bone biomarkers (e.g., alkaline phosphatase/Alpl) and osteogenic transcription factors (e.g., Sp7/Osterix). Furthermore, Suv420h2 is required for matrix mineralization during osteoblast differentiation. We conclude that Suv420h2 controls the H4K20 methylome of osteoblasts and is critical for normal progression of osteoblastogenesis. J. Cell. Biochem. 118: 1262-1272, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Osteoblastos/citología , Osteogénesis , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Línea Celular , Epigénesis Genética , Histonas/metabolismo , Metilación , Ratones , Osteoblastos/metabolismo , Análisis de Secuencia de ARN
4.
J Cell Biochem ; 118(2): 351-360, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27356893

RESUMEN

Osteosarcomas are the most prevalent bone tumors in pediatric patients, but can also occur later in life. Bone tumors have the potential to metastasize to lung and occasionally other vital organs. To understand how osteosarcoma cells interact with their micro-environment to support bone tumor progression and metastasis, we analyzed secreted proteins and exosomes from three human osteosarcoma cell lines. Exosome isolation was validated by transmission electron microscopy (TEM) and immuno-blotting for characteristic biomarkers (CD63, CD9, and CD81). Exosomal and soluble proteins (less than 100 kDa) were identified by mass spectrometry analysis using nanoLC-MS/MS and classified by functional gene ontology clustering. We identified a secretome set of >3,000 proteins for both fractions, and detected proteins that are either common or unique among the three osteosarcoma cell lines. Protein ontology comparison of proteomes from exosomes and exosome-free fractions revealed differences in the enrichment of functional categories associated with different biological processes, including those related to tumor progression (i.e., angiogenesis, cell adhesion, and cell migration). The secretome characteristics of osteosarcoma cells are consistent with the pathological properties of tumor cells with metastatic potential. J. Cell. Biochem. 118: 351-360, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Antígenos CD/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias Óseas/metabolismo , Exosomas/metabolismo , Proteínas de Neoplasias/metabolismo , Osteosarcoma/metabolismo , Línea Celular Tumoral , Humanos
5.
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
6.
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
7.
J Cell Physiol ; 231(5): 1001-14, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26381402

RESUMEN

Epigenetic mechanisms mediate the acquisition of specialized cellular phenotypes during tissue development, maintenance and repair. When phenotype-committed cells transit through mitosis, chromosomal condensation counteracts epigenetic activation of gene expression. Subsequent post-mitotic re-activation of transcription depends on epigenetic DNA and histone modifications, as well as other architecturally bound proteins that "bookmark" the genome. Osteogenic lineage commitment, differentiation and progenitor proliferation require the bone-related runt-related transcription factor Runx2. Here, we characterized a non-genomic mRNA mediated mechanism by which osteoblast precursors retain their phenotype during self-renewal. We show that osteoblasts produce maximal levels of Runx2 mRNA, but not protein, prior to mitotic cell division. Runx2 mRNA partitions symmetrically between daughter cells in a non-chromosomal tubulin-containing compartment. Subsequently, transcription-independent de novo synthesis of Runx2 protein in early G1 phase results in increased functional interactions of Runx2 with a representative osteoblast-specific target gene (osteocalcin/BGLAP2) in chromatin. Somatic transmission of Runx2 mRNAs in osteoblasts and osteosarcoma cells represents a versatile mechanism for translational rather than transcriptional induction of this principal gene regulator to maintain osteoblast phenotype identity after mitosis.


Asunto(s)
Linaje de la Célula/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Patrón de Herencia/genética , Mitosis/genética , Osteoblastos/citología , Osteogénesis/genética , Biosíntesis de Proteínas , Animales , Línea Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Fase G1 , Regulación de la Expresión Génica , Humanos , Interfase , Ratones , Osteoblastos/metabolismo , Osteocalcina/genética , Regiones Promotoras Genéticas , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo
8.
J Biol Chem ; 288(29): 21307-21319, 2013 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-23720736

RESUMEN

Osteosarcoma (OS) is a primary bone tumor that is most prevalent during adolescence. RUNX2, which stimulates differentiation and suppresses proliferation of osteoblasts, is deregulated in OS. Here, we define pathological roles of RUNX2 in the etiology of OS and mechanisms by which RUNX2 expression is stimulated. RUNX2 is often highly expressed in human OS biopsies and cell lines. Small interference RNA-mediated depletion of RUNX2 inhibits growth of U2OS OS cells. RUNX2 levels are inversely linked to loss of p53 (which predisposes to OS) in distinct OS cell lines and osteoblasts. RUNX2 protein levels decrease upon stabilization of p53 with the MDM2 inhibitor Nutlin-3. Elevated RUNX2 protein expression is post-transcriptionally regulated and directly linked to diminished expression of several validated RUNX2 targeting microRNAs in human OS cells compared with mesenchymal progenitor cells. The p53-dependent miR-34c is the most significantly down-regulated RUNX2 targeting microRNAs in OS. Exogenous supplementation of miR-34c markedly decreases RUNX2 protein levels, whereas 3'-UTR reporter assays establish RUNX2 as a direct target of miR-34c in OS cells. Importantly, Nutlin-3-mediated stabilization of p53 increases expression of miR-34c and decreases RUNX2. Thus, a novel p53-miR-34c-RUNX2 network controls cell growth of osseous cells and is compromised in OS.


Asunto(s)
Neoplasias Óseas/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , MicroARNs/metabolismo , Osteosarcoma/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Ciclo Celular/genética , Ciclo Celular/efectos de la radiación , Línea Celular Tumoral , Proliferación Celular/efectos de la radiación , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Daño del ADN , Regulación hacia Abajo/genética , Regulación hacia Abajo/efectos de la radiación , Rayos gamma , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Ratones , Osteosarcoma/genética , Osteosarcoma/patología , Estabilidad Proteica/efectos de la radiación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína p14ARF Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/deficiencia
9.
Vet Parasitol Reg Stud Reports ; 48: 100972, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38316500

RESUMEN

In this study, we report a rare case of tick paralysis in a cat induced by Otobius megnini infestation. An 11-month-old female cat was admitted to a private veterinary clinic in Luling Texas, USA presenting with depression, tachycardia, and flaccid paralysis of the entire body. The four recovered ticks were morphologically and molecularly identified as O. megnini nymphs. Following initial tick removal and treatment with 0.1% milbemycin oxime in the ear canal on the first day of hospitalization, and additional tick removal and topical selamectin treatment on the second day of hospitalization the animal gradually improved. The recovery of the cat after tick removal supports the diagnosis of tick toxicosis. While tick antiserum is not available in North America, prevention of tick infestation and tick-induced paralysis can be effectively accomplished using repellent collars and the compliant use of other ectoparasiticide products year-round.


Asunto(s)
Argasidae , Enfermedades de los Gatos , Parálisis por Garrapatas , Garrapatas , Femenino , Gatos , Animales , Parálisis por Garrapatas/diagnóstico , Parálisis por Garrapatas/tratamiento farmacológico , Parálisis por Garrapatas/veterinaria , Texas , Parálisis/veterinaria , Enfermedades de los Gatos/diagnóstico , Enfermedades de los Gatos/tratamiento farmacológico
10.
J Biol Chem ; 287(7): 4503-17, 2012 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-22158627

RESUMEN

Runt-related transcription factors (RUNX1, RUNX2, and RUNX3) are key lineage-specific regulators of progenitor cell growth and differentiation but also function pathologically as cancer genes that contribute to tumorigenesis. RUNX2 attenuates growth and stimulates maturation of osteoblasts during bone formation but is also robustly expressed in a subset of osteosarcomas, as well as in metastatic breast and prostate tumors. To assess the biological function of RUNX2 in osteosarcoma cells, we examined human genomic promoter interactions for RUNX2 using chromatin immunoprecipitation (ChIP)-microarray analysis in SAOS-2 cells. Promoter binding of both RUNX2 and RNA polymerase II was compared with gene expression profiles of cells in which RUNX2 was depleted by RNA interference. Many RUNX2-bound loci (1550 of 2339 total) exhibit promoter occupancy by RNA polymerase II and contain the RUNX consensus motif 5'-((T/A/C)G(T/A/C)GG(T/G). Gene ontology analysis indicates that RUNX2 controls components of multiple signaling pathways (e.g. WNT, TGFß, TNFα, and interleukins), as well as genes linked to cell motility and adhesion (e.g. the focal adhesion-related genes FAK/PTK2 and TLN1). Our results reveal that siRNA depletion of RUNX2, PTK2, or TLN1 diminishes motility of U2OS osteosarcoma cells. Thus, RUNX2 binding to diverse gene loci may support the biological properties of osteosarcoma cells.


Asunto(s)
Neoplasias Óseas/metabolismo , Movimiento Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Genoma Humano , Proteínas de Neoplasias/metabolismo , Osteosarcoma/metabolismo , Elementos de Respuesta , Neoplasias Óseas/genética , Adhesión Celular/genética , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Sitios Genéticos , Humanos , Proteínas de Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteosarcoma/genética , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo
11.
J Cell Physiol ; 228(4): 714-23, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22949168

RESUMEN

Runx2 regulates osteogenic differentiation and bone formation, but also suppresses pre-osteoblast proliferation by affecting cell cycle progression in the G(1) phase. The growth suppressive potential of Runx2 is normally inactivated in part by protein destabilization, which permits cell cycle progression beyond the G(1)/S phase transition, and Runx2 is again up-regulated after mitosis. Runx2 expression also correlates with metastasis and poor chemotherapy response in osteosarcoma. Here we show that six human osteosarcoma cell lines (SaOS, MG63, U2OS, HOS, G292, and 143B) have different growth rates, which is consistent with differences in the lengths of the cell cycle. Runx2 protein levels are cell cycle-regulated with respect to the G(1)/S phase transition in U2OS, HOS, G292, and 143B cells. In contrast, Runx2 protein levels are constitutively expressed during the cell cycle in SaOS and MG63 cells. Forced expression of Runx2 suppresses growth in all cell lines indicating that accumulation of Runx2 in excess of its pre-established levels in a given cell type triggers one or more anti-proliferative pathways in osteosarcoma cells. Thus, regulatory mechanisms controlling Runx2 expression in osteosarcoma cells must balance Runx2 protein levels to promote its putative oncogenic functions, while avoiding suppression of bone tumor growth.


Asunto(s)
Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Osteosarcoma/genética , Osteosarcoma/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Neoplasias Óseas/patología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Fase G1/genética , Humanos , Osteosarcoma/patología , Fase S/genética
12.
Nature ; 445(7126): 442-6, 2007 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-17251981

RESUMEN

Regulation of ribosomal RNA genes is a fundamental process that supports the growth of cells and is tightly coupled with cell differentiation. Although rRNA transcriptional control by RNA polymerase I (Pol I) and associated factors is well studied, the lineage-specific mechanisms governing rRNA expression remain elusive. Runt-related transcription factors Runx1, Runx2 and Runx3 establish and maintain cell identity, and convey phenotypic information through successive cell divisions for regulatory events that determine cell cycle progression or exit in progeny cells. Here we establish that mammalian Runx2 not only controls lineage commitment and cell proliferation by regulating genes transcribed by RNA Pol II, but also acts as a repressor of RNA Pol I mediated rRNA synthesis. Within the condensed mitotic chromosomes we find that Runx2 is retained in large discrete foci at nucleolar organizing regions where rRNA genes reside. These Runx2 chromosomal foci are associated with open chromatin, co-localize with the RNA Pol I transcription factor UBF1, and undergo transition into nucleoli at sites of rRNA synthesis during interphase. Ribosomal RNA transcription and protein synthesis are enhanced by Runx2 deficiency that results from gene ablation or RNA interference, whereas induction of Runx2 specifically and directly represses rDNA promoter activity. Runx2 forms complexes containing the RNA Pol I transcription factors UBF1 and SL1, co-occupies the rRNA gene promoter with these factors in vivo, and affects local chromatin histone modifications at rDNA regulatory regions. Thus Runx2 is a critical mechanistic link between cell fate, proliferation and growth control. Our results suggest that lineage-specific control of ribosomal biogenesis may be a fundamental function of transcription factors that govern cell fate.


Asunto(s)
Linaje de la Célula , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Genes de ARNr/genética , Mitosis , Transcripción Genética , Animales , Secuencia de Bases , Cromátides/genética , Cromátides/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/deficiencia , ADN Ribosómico/genética , Humanos , Interfase , Metafase , Ratones , Mitosis/genética , Modelos Biológicos , Complejos Multienzimáticos/metabolismo , Proteínas del Complejo de Iniciación de Transcripción Pol1/metabolismo , ARN Polimerasa I/metabolismo , ARN Ribosómico/biosíntesis , Proteínas Represoras/metabolismo , Transcripción Genética/genética
14.
J Cell Physiol ; 226(7): 1953-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21506126

RESUMEN

ß-Catenin is a key protein in the canonical Wnt signaling pathway and in many cancers alterations in transcriptional activity of its components are observed. This pathway is up-regulated by the protein kinase CK2, but the underlying mechanism of this change is unknown. It has been demonstrated that CK2 hyperactivates AKT/PKB by phosphorylation at Ser129, and AKT phosphorylates ß-catenin at Ser552, which in turn, promotes its nuclear localization and transcriptional activity. However, the consequences of CK2-dependent hyperactivation of AKT on ß-catenin activity and cell viability have not been evaluated. We assessed this regulatory process by manipulating the activity of CK2 and AKT through overexpression of wild-type, constitutively active and dominant negative forms of these proteins as well as analyzing ß-catenin-dependent transcriptional activity, survivin expression and viability in HEK-293T cells. We observed that CK2α overexpression up-regulated the ß-catenin transcriptional activity, which correlated to an increased nuclear localization of ß-catenin as well as survivin expression. Importantly, these effects were strongly reversed when an AKT-S129A mutant was co-expressed in the same cells, followed by a significant decrease in cell viability but no changes in ß-catenin stability. Taken together, the data suggest that the CK2α-dependent up-regulation of ß-catenin activity requires phosphorylation of AKT in human embryonic kidney cells.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transcripción Genética , beta Catenina/metabolismo , Quinasa de la Caseína II/genética , Supervivencia Celular , Células HEK293 , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Mutación , Fosforilación , Proteínas Recombinantes de Fusión/metabolismo , Serina , Survivin , Transfección , Regulación hacia Arriba , beta Catenina/genética
15.
Mol Cell Biochem ; 356(1-2): 127-32, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21735093

RESUMEN

ß-Catenin is crucial in the canonical Wnt signaling pathway. This pathway is up-regulated by CK2 which is associated with an enhanced expression of the antiapoptotic protein survivin, although the underlying molecular mechanism is unknown. AKT/PKB kinase phosphorylates and promotes ß-catenin transcriptional activity, whereas CK2 hyperactivates AKT by phosphorylation at Ser129; however, the role of this phosphorylation on ß-catenin transcriptional activity and cell survival is unclear. We studied in HEK-293T cells, the effect of CK2-dependent hyperactivation of AKT on cell viability, as well as analyzed ß-catenin subcellular localization and transcriptional activity and survivin expression. CK2α overexpression led to an augmented ß-catenin-dependent transcription and protein levels of survivin, and consequently an enhanced resistance to apoptosis. However, CK2α-enhancing effects were reversed when an AKT mutant deficient in Ser129 phosphorylation by CK2 was co-expressed. Therefore, our results strongly suggest that CK2α-specific enhancement of ß-catenin transcriptional activity as well as cell survival may depend on AKT hyperactivation by CK2.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Proteínas Inhibidoras de la Apoptosis/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , beta Catenina/metabolismo , Apoptosis , Núcleo Celular/metabolismo , Supervivencia Celular , Células HEK293 , Humanos , Proteínas Mutantes/metabolismo , Fosforilación , Unión Proteica , Transporte de Proteínas , Regulación hacia Arriba
16.
Front Immunol ; 12: 664756, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34012448

RESUMEN

Periodontitis is a chronic inflammatory disease associated with the formation of dysbiotic plaque biofilms and characterized by the progressive destruction of the alveolar bone. The transition from health to disease is characterized by a shift in periodontal immune cell composition, from mostly innate (neutrophils) to adaptive (T lymphocytes) immune responses. Resolvin E1 (RvE1) is a specialized pro-resolution mediator (SPMs), produced in response to inflammation, to enhance its resolution. Previous studies have indicated the therapeutic potential of RvE1 in periodontal disease; however, the impact of RvE1 in the microbial-elicited osteoclastogenic immune response remains uncharacterized in vivo. In the present study, we studied the impact of RvE1 on the gingival inflammatory infiltrate formation during periodontitis in a mouse model. First, we characterized the temporal-dependent changes of the main immune cells infiltrating the gingiva by flow cytometry. Then, we evaluated the impact of early or delayed RvE1 administration on the gingival immune infiltration and cervical lymph nodes composition. We observed a consistent inhibitory outcome on T cells -particularly effector T cells- and a protective effect on regulatory T cells (Tregs). Our data further demonstrated the wide range of actions of RvE1, its preventive role in the establishment of the adaptive immune response during inflammation, and bone protective capacity.


Asunto(s)
Ácido Eicosapentaenoico/análogos & derivados , Gingivitis/etiología , Gingivitis/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Pérdida de Hueso Alveolar/etiología , Pérdida de Hueso Alveolar/metabolismo , Pérdida de Hueso Alveolar/patología , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ácido Eicosapentaenoico/farmacología , Gingivitis/tratamiento farmacológico , Gingivitis/patología , Inmunofenotipificación , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Ratones , Neutrófilos/inmunología , Neutrófilos/metabolismo , Periodontitis/etiología , Periodontitis/metabolismo , Periodontitis/patología , Linfocitos T/patología
17.
Mol Biol Rep ; 37(6): 2927-33, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19842061

RESUMEN

Toxoplasma gondii is one of the most successful protozoan parasites given its ability to manipulate the immune system and establish a chronic infection. It is a parasite with a significant impact on human health, mainly in immunocompromised patients. In Europe and North America, only a few clonal genotypes (I, II and III) seem to be responsible for the vast majority of Toxoplasma infections. Surface antigen 2 gene (SAG2) has been extensively used for genotyping T. gondii isolates. The analysis of this locus reveals that in Northern hemisphere, human disease causing isolates are mainly type II, whereas T. gondii isolated from different animals are both type II and III. Since the immune response depends on parasite genotype, it seems relevant to characterize parasites producing human toxoplasmosis in different geographical areas. The growing information about the prevalent T. gondii genotypes in South America mostly refers to domestic animals. This is the first report of genetic characterization of T. gondii isolates from clinical samples in Chile, South America. All the samples analyzed corresponded to SAG2 type I isolates, and they differ from classic SAG2 type I by genetic polymorphisms. This study contributes to the scarce available information on T. gondii at South America, and reinforces an emerging concept suggesting that SAG2 type I, rather than II, parasites are a frequent cause of clinical toxoplasmosis in this continent.


Asunto(s)
Antígenos de Protozoos/genética , Proteínas Protozoarias/genética , Toxoplasma/genética , Toxoplasmosis/genética , Toxoplasmosis/parasitología , Cromosomas/genética , Electroforesis en Gel de Campo Pulsado , Sitios Genéticos/genética , Genotipo , Humanos , Polimorfismo de Nucleótido Simple/genética , Técnica del ADN Polimorfo Amplificado Aleatorio , Análisis de Secuencia de ADN , Toxoplasma/aislamiento & purificación
18.
Sci Rep ; 10(1): 19018, 2020 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-33149125

RESUMEN

The alveolar bone resorption is a distinctive feature of periodontitis progression and determinant for tooth loss. Regulatory T lymphocytes (Tregs) display immuno-suppressive mechanisms and tissue repairing functions, which are critical to support periodontal health. Tregs may become unstable and dysfunctional under inflammatory conditions, which can even accelerate tissue destruction. In this study, experimental periodontitis was associated with the progressive and increased presence of Th17 and Treg-related mediators in the gingiva (IL-6, IL-17A, IL-17F, RANKL, IL-10, TGF-ß and GITR; P < 0.05), and the proliferation of both Treg and Th17 cells in cervical lymph nodes. Tregs from cervical lymph nodes had reduced Foxp3 expression (> 25% MFI loss) and increased IL-17A expression (> 15%), compared with Tregs from spleen and healthy controls. Tregs gene expression analysis showed a differential signature between health and disease, with increased expression of Th17-associated factors in periodontitis-derived Tregs. The ex vivo suppression capacity of Tregs on osteoclastic differentiation was significantly lower in Tregs obtained from periodontally diseased animals compared to controls (P < 0.05), as identified by the increased number of TRAP+ osteoclasts (P < 0.01) in the Tregs/pre-osteoclast co-cultures. Taken together, these results demonstrate that Tregs become phenotypically unstable and lose anti-osteoclastogenic properties during experimental periodontitis; thus, further promoting the Th17-driven bone loss.


Asunto(s)
Osteogénesis/inmunología , Periodontitis/inmunología , Linfocitos T Reguladores/inmunología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/patología , Enfermedad Crónica , Técnicas de Cocultivo , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Expresión Génica , Inmunofenotipificación , Interleucina-17/biosíntesis , Ganglios Linfáticos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Cuello , Linfocitos T Reguladores/patología
19.
J Cell Physiol ; 221(3): 560-71, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19739101

RESUMEN

Bone formation and osteoblast differentiation require the functional expression of the Runx2/Cbfbeta heterodimeric transcription factor complex. Runx2 is also a suppressor of proliferation in osteoblasts by attenuating cell cycle progression in G(1). Runx2 levels are modulated during the cell cycle, which are maximal in G(1) and minimal beyond the G(1)/S phase transition (S, G(2), and M phases). It is not known whether Cbfbeta gene expression is cell cycle controlled in preosteoblasts nor how Runx2 or Cbfbeta are regulated during the cell cycle in bone cancer cells. We investigated Runx2 and Cbfbeta gene expression during cell cycle progression in MC3T3-E1 osteoblasts, as well as ROS17/2.8 and SaOS-2 osteosarcoma cells. Runx2 protein levels are reduced as expected in MC3T3-E1 cells arrested in late G(1) (by mimosine) or M phase (by nocodazole), but not in cell cycle arrested osteosarcoma cells. Cbfbeta protein levels are cell cycle independent in both osteoblasts and osteosarcoma cells. In synchronized MC3T3-E1 osteoblasts progressing from late G1 or mitosis, Runx2 levels but not Cbfbeta levels are cell cycle regulated. However, both factors are constitutively elevated throughout the cell cycle in osteosarcoma cells. Proteasome inhibition by MG132 stabilizes Runx2 protein levels in late G(1) and S in MC3T3-E1 cells, but not in ROS17/2.8 and SaOS-2 osteosarcoma cells. Thus, proteasomal degradation of Runx2 is deregulated in osteosarcoma cells. We propose that cell cycle control of Runx2 gene expression is impaired in osteosarcomas and that this deregulation may contribute to the pathogenesis of osteosarcoma.


Asunto(s)
Ciclo Celular/fisiología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Osteosarcoma/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad beta del Factor de Unión al Sitio Principal/genética , Inhibidores de Cisteína Proteinasa , Fase G1/fisiología , Expresión Génica/genética , Humanos , Leupeptinas/farmacología , Ratones , Mitosis/fisiología , Osteoblastos/citología , Osteoblastos/metabolismo , Osteosarcoma/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Ratas , Ubiquitinación/efectos de los fármacos
20.
J Cell Physiol ; 221(3): 778-88, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19746444

RESUMEN

Osteosarcomas are the most prevalent primary bone tumors found in pediatric patients. To understand their molecular etiology, cell culture models are used to define disease mechanisms under controlled conditions. Many osteosarcoma cell lines (e.g., SAOS-2, U2OS, MG63) are derived from Caucasian patients. However, patients exhibit individual and ethnic differences in their responsiveness to irradiation and chemotherapy. This motivated the establishment of osteosarcoma cell lines (OS1, OS2, OS3) from three ethnically Chinese patients. OS1 cells, derived from a pre-chemotherapeutic tumor in the femur of a 6-year-old female, were examined for molecular markers characteristic for osteoblasts, stem cells, and cell cycle control by immunohistochemistry, reverse transcriptase-PCR, Western blotting and flow cytometry. OS1 have aberrant G-banded karyotypes, possibly reflecting chromosomal abnormalities related to p53 deficiency. OS1 had ossification profiles similar to human fetal osteoblasts rather than SAOS-2 which ossifies ab initio (P < 0.05). Absence of p53 correlates with increased Runx2 expression, while the slow proliferation of OS1 cells is perhaps attenuated by pRB retention. OS1 express mesenchymal stem cell markers (CD44, CD105) and differ in relative expression of CD29, CD63, and CD71 to SAOS-2. (P < 0.05). Cell cycle synchronization with nocodazole did not affect Runx2 and CDK1 levels but decreased cyclin-E and increased cyclin-A (P < 0.05). Xenotransplantion of OS1 in SCID mice yields spontaneous tumors that were larger and grew faster than SAOS-2 transplants. Hence, OS1 is a new osteosarcoma cell culture model derived from a pre-chemotherapeutic ethnic Chinese patient, for mechanistic studies and development of therapeutic strategies to counteract metastasis and deregulation of mesenchymal development.


Asunto(s)
Diferenciación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Osteoblastos/patología , Osteosarcoma/metabolismo , Osteosarcoma/patología , Proteína de Retinoblastoma/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Adolescente , Animales , Antígenos CD/metabolismo , Pueblo Asiatico , Calcificación Fisiológica/fisiología , Ciclo Celular/efectos de los fármacos , Desdiferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Niño , Aberraciones Cromosómicas , Colágeno Tipo I/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Ciclinas/genética , Ciclinas/metabolismo , Femenino , Expresión Génica/genética , Humanos , Antígeno Ki-67/metabolismo , Ratones , Ratones SCID , Nocodazol/farmacología , Osteocalcina/metabolismo , Osteosarcoma/diagnóstico , Osteosarcoma/genética , Proteína de Retinoblastoma/genética , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA