Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Bone ; 181: 117014, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38218304

RESUMEN

Deletion of Runx2 gene in proliferating chondrocytes results in complete failure of endochondral ossification and perinatal lethality. We reported recently that mice with Runx2 deletion specifically in hypertrophic chondrocytes (HCs) using the Col10a1-Cre transgene survive and exhibit enlarged growth plates due to decreased HC apoptosis and cartilage resorption. Bulk of chondrogenesis occurs postnatally, however, the role of Runx2 in HCs during postnatal chondrogenesis is unknown. Despite limb dwarfism, adult homozygous (Runx2HC/HC) mice showed a significant increase in length of growth plate and articular cartilage. Consistent with doubling of the hypertrophic zone, collagen type X expression was increased in Runx2HC/HC mice. In sharp contrast, expression of metalloproteinases and aggrecanases were markedly decreased. Impaired cartilage degradation was evident by the retention of significant amount of safranin-O positive cartilage. Histomorphometry and µCT uncovered increased trabecular bone mass with a significant increase in BV/TV ratio, trabecular number, thickness, and a decrease in trabecular space in Runx2HC/HC mice. To identify if this is due to increased bone synthesis, expression of osteoblast differentiation markers was evaluated and found to be comparable amongst littermates. Histomorphometry confirmed similar number of osteoblasts in the littermates. Furthermore, dynamic bone synthesis showed no differences in mineral apposition or bone formation rates. Surprisingly, three-point-bending test revealed Runx2HC/HC bones to be structurally less strong. Interestingly, both the number and surface of osteoclasts were markedly reduced in Runx2HC/HC littermates. Rankl and IL-17a ligands that promote osteoclast differentiation were markedly reduced in Runx2HC/HC mice. Bone marrow cultures were performed to independently establish Runx2 and hypertrophic chondrocytes role in osteoclast development. The culture from the Runx2HC/HC mice formed significantly fewer and smaller osteoclasts. The expression of mature osteoclast markers, Ctsk and Mmp9, were significantly reduced in the cultures from Runx2HC/HC mice. Thus, Runx2 functions extend beyond embryonic development and chondrocyte hypertrophy by regulating cartilage degradation, osteoclast differentiation, and bone resorption during postnatal endochondral ossification.


Asunto(s)
Resorción Ósea , Condrocitos , Animales , Ratones , Condrocitos/metabolismo , Osteoclastos/metabolismo , Cartílago/metabolismo , Osteogénesis/fisiología , Resorción Ósea/metabolismo , Hipertrofia , Diferenciación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo
2.
Front Immunol ; 13: 906649, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36189270

RESUMEN

The early B cell protein λ5 is an essential component of the surrogate light chain and the preB cell receptor (preBCR), which is critical for optimal B cell development. To investigate the effect of λ5 and/or B cells on bone acquisition over time, we developed a panel of JH -/- , λ5-/-, JH -/- λ5-/-, and wild-type (WT) BALB/c mice and then studied postnatal bone development and aging in these mice at one, six, twelve, and twenty-two months of age. The trabecular bone volume over total volume (BV/TV) in JH -/- mice was similar to WT mice at all ages. In contrast, at six months of age and thereafter, λ5-/- and JH -/- λ5-/- mice demonstrated a severe decrease in trabecular bone mass. Surprisingly, bone mass in six-month-old λ5-/- and JH -/- λ5-/- mice was similar to or even lower than in aged (twenty-two-months) WT mice, suggesting accelerated skeletal aging. The postnatal development and the acquisition of cortical bone mass in JH -/- λ5-/- mice were generally comparable to WT. However, JH -/- λ5-/- mice showed a significant decrease in cortical BV/TV at six- and twelve months of age. To examine the contribution of λ5 and B cells to postnatal bone synthesis, we separately transplanted whole bone marrow cells from JH -/- λ5-/- and WT mice into irradiated JH -/- λ5-/- and WT recipients. WT recipients of JH -/- λ5-/- marrow cells failed to show acquisition of trabecular bone mass, whereas transplanting WT marrow cells into JH -/- λ5-/- recipients led to the recovery of trabecular bone mass. Transfer of WT marrow cells into JH -/- λ5-/- mice promoted synthesis of new cortical and trabecular bone. Our findings indicate that λ5 plays a major role in preserving bone mass during postnatal development and skeletal aging which is distinct from its role in B cell development. The absence of both λ5 and B cells in JH -/- λ5-/- mice leads to delayed acquisition of cortical bone during postnatal development. Dissecting the mechanism(s) by which λ5 regulates bone homeostasis may provide new avenues for the treatment of age-related loss of bone mass and osteoporosis.


Asunto(s)
Linfocitos B , Receptores de Células Precursoras de Linfocitos B , Envejecimiento , Animales , Linfocitos B/metabolismo , Densidad Ósea , Inmunoglobulina de Cadenas Ligeras Subrogadas/metabolismo , Ratones , Ratones Endogámicos BALB C , Receptores de Células Precursoras de Linfocitos B/metabolismo
3.
Mol Cancer Ther ; 21(2): 347-358, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34907087

RESUMEN

Multiple myeloma is a plasma cell malignancy that thrives in the bone marrow (BM). The proteasome inhibitor bortezomib is one of the most effective first-line chemotherapeutic drugs for multiple myeloma; however, 15% to 20% of high-risk patients do not respond to or become resistant to this drug and the mechanisms of chemoresistance remain unclear. We previously demonstrated that multiple myeloma cells inhibit Runt-related transcription factor 2 (Runx2) in pre- and immature osteoblasts (OB), and that this OB-Runx2 deficiency induces a cytokine-rich and immunosuppressive microenvironment in the BM. In the current study, we assessed the impact of OB-Runx2 deficiency on the outcome of bortezomib treatment using OB-Runx2+/+ and OB-Runx2-/- mouse models of multiple myeloma. In vitro and in vivo experiments revealed that OB-Runx2 deficiency induces multiple myeloma cell resistance to bortezomib via the upregulation of immunosuppressive myeloid-derived suppressor cells (MDSCs), downregulation of cytotoxic T cells, and activation of TGFß1 in the BM. In multiple myeloma tumor-bearing OB-Runx2-/- mice, treatment with SRI31277, an antagonist of thrombospondin-1 (TSP-1)-mediated TGFß1 activation, reversed the BM immunosuppression and significantly reduced tumor burden. Furthermore, treatment with SRI31277 combined with bortezomib alleviated multiple myeloma cell resistance to bortezomib-induced apoptosis caused by OB-Runx2 deficiency in cocultured cells and produced a synergistic effect on tumor burden in OB-Runx2-/- mice. Depletion of MDSCs by 5-fluorouracil or gemcitabine similarly reversed the immunosuppressive effects and bortezomib resistance induced by OB-Runx2 deficiency in tumor-bearing mice, indicating the importance of the immune environment for drug resistance and suggesting new strategies to overcome bortezomib resistance in the treatment of multiple myeloma.


Asunto(s)
Médula Ósea/metabolismo , Bortezomib/uso terapéutico , Subunidad alfa 1 del Factor de Unión al Sitio Principal/deficiencia , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Osteoblastos/metabolismo , Trombospondina 1/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Bortezomib/farmacología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Ratones , Mieloma Múltiple/patología
4.
Front Immunol ; 13: 1114732, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36861066

RESUMEN

[This corrects the article DOI: 10.3389/fimmu.2022.906649.].

5.
Matrix Biol Plus ; 12: 100088, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34805821

RESUMEN

The RUNX2 transcription factor is a key regulator for the development of cartilage and bone. Global or resting chondrocyte-specific deletion of the Runx2 gene results in failure of chondrocyte hypertrophy, endochondral ossification, and perinatal lethality. The terminally mature hypertrophic chondrocyte regulates critical steps of endochondral ossification. Importantly, expression of the Runx2 gene starts in the resting chondrocyte and increases progressively, reaching the maximum level in hypertrophic chondrocytes. However, the RUNX2 role after chondrocyte hypertrophy remains unknown. To answer this question, we deleted the Runx2 gene specifically in hypertrophic chondrocytes using the Col10-Cre line. Mice lacking the Runx2 gene in hypertrophic chondrocytes (Runx2HC/HC ) survive but exhibit limb dwarfism. Interestingly, the length of the hypertrophic chondrocyte zone is doubled in the growth plate of Runx2HC/HC mice. Expression of pro-apoptotic Bax decreased significantly while anti-apoptotic Bcl2 remains unchanged leading to a four-fold increase in the Bcl2/Bax ratio in mutant mice. In line with this, a significant reduction in apoptosis of Runx2HC/HC hypertrophic chondrocyte is noted. A large amount of cartilage matrix is present in the long bones that extend toward the diaphyseal region of Runx2HC/HC mice. This is not due to enhanced synthesis of the cartilage matrix as the expression of both collagen type 2 and aggrecan were comparable among Runx2HC/HC and WT littermates. Our qPCR analysis demonstrates the increased amount of cartilage matrix is due to impaired expression of cartilage degrading enzymes such as metalloproteinase and aggrecanase as well as tissue inhibitor of metalloproteinases. Moreover, a significant decrease of TRAP positive chondroclasts was noted along the cartilage islands in Runx2HC/HC mice. Consistently, qPCR data showed an 81% reduction in the Rankl/Opg ratio in Runx2HC/HC littermates, which is inhibitory for chondroclast differentiation. Finally, we assess if increase cartilage matrix in Runx2HC/HC mice serves as a template for bone and mineral deposition using micro-CT and Von Kossa. The mutant mice exhibit a significant increase in trabecular bone mass compared to littermates. In summary, our findings have uncovered a novel role of Runx2 in apoptosis of hypertrophic chondrocytes and degradation of cartilage matrix during endochondral ossification.

6.
Cancer Res ; 80(5): 1036-1048, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31911552

RESUMEN

Multiple myeloma is a plasma cell malignancy that thrives in the bone marrow (BM), with frequent progression to new local and distant bone sites. Our previous studies demonstrated that multiple myeloma cells at primary sites secrete soluble factors and suppress osteoblastogenesis via the inhibition of Runt-related transcription factor 2 (Runx2) in pre- and immature osteoblasts (OB) in new bone sites, prior to the arrival of metastatic tumor cells. However, it is unknown whether OB-Runx2 suppression in new bone sites feeds back to promote multiple myeloma dissemination to and progression in these areas. Hence, we developed a syngeneic mouse model of multiple myeloma in which Runx2 is specifically deleted in the immature OBs of C57BL6/KaLwRij mice (OB-Runx2-/- mice) to study the effect of OB-Runx2 deficiency on multiple myeloma progression in new bone sites. In vivo studies with this model demonstrated that OB-Runx2 deficiency attracts multiple myeloma cells and promotes multiple myeloma tumor growth in bone. Mechanistic studies further revealed that OB-Runx2 deficiency induces an immunosuppressive microenvironment in BM that is marked by an increase in the concentration and activation of myeloid-derived suppressor cells (MDSC) and the suppression and exhaustion of cytotoxic CD8+ T cells. In contrast, MDSC depletion by either gemcitabine or 5-fluorouracil treatment in OB-Runx2-/- mice prevented these effects and inhibited multiple myeloma tumor growth in BM. These novel discoveries demonstrate that OB-Runx2 deficiency in new bone sites promotes multiple myeloma dissemination and progression by increasing metastatic cytokines and MDSCs in BM and inhibiting BM immunity. Importantly, MDSC depletion can block multiple myeloma progression promoted by OB-Runx2 deficiency.Significance: This study demonstrates that Runx2 deficiency in immature osteoblasts at distant bone sites attracts myeloma cells and allows myeloma progression in new bone sites via OB-secreted metastatic cytokines and MDSC-mediated suppression of bone marrow immunity.


Asunto(s)
Neoplasias Óseas/secundario , Subunidad alfa 1 del Factor de Unión al Sitio Principal/deficiencia , Mieloma Múltiple/patología , Osteoblastos/patología , Microambiente Tumoral/inmunología , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/inmunología , Médula Ósea/patología , Neoplasias Óseas/inmunología , Huesos/citología , Huesos/efectos de los fármacos , Huesos/inmunología , Huesos/patología , Línea Celular Tumoral/trasplante , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Modelos Animales de Enfermedad , Femenino , Fluorouracilo , Humanos , Masculino , Ratones , Ratones Noqueados , Mieloma Múltiple/inmunología , Células Supresoras de Origen Mieloide/efectos de los fármacos , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/patología , Osteoblastos/inmunología , Gemcitabina
7.
Front Immunol ; 10: 2063, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31552025

RESUMEN

In the bone marrow, preB cells are found adjacent to the bone endosteum where bone synthesizing osteoblast and bone resorbing osteoclasts reside. Although there is evidence of interactions between preB and bone cells, the factors that contribute to such interactions are poorly understood. A critical checkpoint for preB cell development assesses the integrity of the nascent immunoglobulin µ heavy chain (HC) by testing whether it can participate in the formation of a preB cell receptor (preBCR), composed of the µ HC and surrogate light chain (LC). In this work, we tested whether loss of preBCR components can affect bone synthesis. A panel of gene targeted mice with sequential blocks in preBCR formation or function [surrogate light chain component lambda 5 deleted (λ5-/-), transmembrane domain of µHC deleted (IgM-mem-/-), and CD19 preBCR co-receptor deleted (CD19-/-)] were evaluated for effects on postnatal bone synthesis. Postnatal bone mass was analyzed in 6 month old mice using µ-CT, histomorphometry and double calcein labeling. Both cortical and trabecular bone mass were significantly decreased in the femurs of the λ5 and IgM-mem deficient mice. Histomorphometric analysis showed a decrease in the numbers of osteoblasts and osteoclasts in all three mutant strains. Double calcein labeling revealed a significant decrease in dynamic synthesis and mineralization of bone in λ5-/- mice. Our data strongly suggest that interference with preBCR formation or function affects bone homeostasis independent of the presence or absence of mature B cells, and that components of the preBCR play important, and potentially distinct, roles in regulating adult bone mass.


Asunto(s)
Huesos/inmunología , Cadenas Pesadas de Inmunoglobulina/inmunología , Inmunoglobulina de Cadenas Ligeras Subrogadas/inmunología , Cadenas mu de Inmunoglobulina/inmunología , Receptores de Células Precursoras de Linfocitos B/inmunología , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Huesos/diagnóstico por imagen , Huesos/metabolismo , Fémur/diagnóstico por imagen , Fémur/inmunología , Fémur/metabolismo , Homeostasis/inmunología , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/metabolismo , Inmunoglobulina de Cadenas Ligeras Subrogadas/genética , Inmunoglobulina de Cadenas Ligeras Subrogadas/metabolismo , Cadenas mu de Inmunoglobulina/genética , Cadenas mu de Inmunoglobulina/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/citología , Osteoblastos/inmunología , Osteoblastos/metabolismo , Osteoclastos/citología , Osteoclastos/inmunología , Osteoclastos/metabolismo , Receptores de Células Precursoras de Linfocitos B/genética , Receptores de Células Precursoras de Linfocitos B/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Microtomografía por Rayos X/métodos
8.
Sci Rep ; 8(1): 15749, 2018 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-30356078

RESUMEN

To date, bone tissue regeneration strategies lack an approach that effectively provides an osteogenic and angiogenic environment conducive to bone growth. In the current study, we evaluated the osteogenic and angiogenic response of human mesenchymal stem cells (hMSCs) and green fluorescent protein-expressing human umbilical vein endothelial cells (GFP-HUVECs) cocultured on a self-assembled, peptide amphiphile nanomatrix functionalized with the cell adhesive ligand RGDS (PA-RGDS). Analysis of alkaline phosphatase activity, von Kossa staining, Alizarin Red quantification, and osteogenic gene expression, indicates a significant synergistic effect between the PA-RGDS nanomatrix and coculture that promoted hMSC osteogenesis. In addition, coculturing on PA-RGDS resulted in enhanced HUVEC network formation and upregulated vascular endothelial growth factor gene and protein expression. Though PA-RGDS and coculturing hMSCs with HUVECs were each previously reported to individually enhance hMSC osteogenesis, this study is the first to demonstrate a synergistic promotion of HUVEC angiogenesis and hMSC osteogenesis by integrating coculturing with the PA-RGDS nanomatrix. We believe that using the combination of hMSC/HUVEC coculture and PA-RGDS substrate is an efficient method for promoting osteogenesis and angiogenesis, which has immense potential as an efficacious, engineered platform for bone tissue regeneration.


Asunto(s)
Técnicas de Cocultivo/métodos , Células Endoteliales de la Vena Umbilical Humana/citología , Células Madre Mesenquimatosas/citología , Neovascularización Fisiológica , Osteogénesis , Medicina Regenerativa/métodos , Regeneración Ósea , Células Cultivadas , Humanos , Oligopéptidos/farmacología
9.
Connect Tissue Res ; 59(sup1): 52-54, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29745807

RESUMEN

Current studies offer little insight on how epigenetic remodeling of bone-specific chromatin maintains bone mass in vivo. Understanding this gap and precise mechanism is pivotal for future therapeutic innovation to prevent bone loss. Recently, we found that low bone mass is associated with decreased H3K27 acetylation (activating histone modification) of bone specific gene promoters. Here, we aim to elucidate the epigenetic mechanisms by which a miRNA cluster controls bone synthesis and homeostasis by regulating chromatin accessibility and H3K27 acetylation. In order to decipher the epigenetic axis that regulates osteogenesis, we studied a drug inducible anti-miR-23a cluster (miR-23a ClZIP) knockdown mouse model. MiR-23a cluster knockdown (heterozygous) mice developed high bone mass. These mice displayed increased expression of Runx2 and Baf45a, essential factors for skeletogenesis; and decreased expression of Ezh2, a chromatin repressor indispensable for skeletogenesis. ChIP assays using miR-23a Cl knockdown calvarial cells revealed a BAF45A-EZH2 epigenetic antagonistic mechanism that maintains bone formation. Together, our findings support that the miR-23a Cl connection with tissue-specific RUNX2-BAF45A-EZH2 function is a novel molecular epigenetic axis through which a miRNA cluster orchestrates chromatin modification to elicit major effects on osteogenesis in vivo.


Asunto(s)
Diferenciación Celular/fisiología , Epigénesis Genética/fisiología , Osteoblastos/metabolismo , Osteogénesis/fisiología , Acetilación , Animales , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Histonas/genética , Histonas/metabolismo , Ratones , Ratones Transgénicos , MicroARNs/genética , MicroARNs/metabolismo , Osteoblastos/citología
10.
J Bone Miner Res ; 33(6): 1126-1140, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29405385

RESUMEN

The Sp7/Osterix transcription factor is essential for bone development. Mutations of the Sp7 gene in humans are associated with craniofacial anomalies and osteogenesis imperfecta. However, the role of Sp7 in embryonic tooth development remains unknown. Here we identified the functional requirement of Sp7 for dentin synthesis and tooth development. Sp7-null mice exhibit craniofacial dysmorphogenesis and are completely void of alveolar bone. Surprisingly, initial tooth morphogenesis progressed normally in Sp7-null mice. Thus the formation of alveolar bone is not a prerequisite for tooth morphogenesis. Sp7 is required for mineralization of palatal tissue but is not essential for palatal fusion. The reduced proliferative capacity of Sp7-deficient ectomesenchyme results in small and misshapen teeth with randomly arranged cuboidal preodontoblasts and preameloblasts. Sp7 promotes functional maturation and polarization of odontoblasts. Markers of mature odontoblast (Col1a, Oc, Dspp, Dmp1) and ameloblast (Enam, Amelx, Mmp20, Amtn, Klk4) are barely expressed in incisors and molar tissues of Sp7-null mice. Consequently, dentin and enamel matrix are absent in the Sp7-null littermates. Interestingly, the Sp7 expression is restricted to cells of the dental mesenchyme indicating the effect on oral epithelium-derived ameloblasts is cell-nonautonomous. Abundant expression of Fgf3 and Fgf8 ligand was noted in the developing tooth of wild-type mice. Both ligands were remarkably absent in the Sp7-null incisor and molar, suggesting cross-signaling between mesenchyme and epithelium is disrupted. Finally, promoter-reporter assays revealed that Sp7 directly controls the expression of Fgf-ligands. Together, our data demonstrate that Sp7 is obligatory for the differentiation of both ameloblasts and odontoblasts but not for the initial tooth morphogenesis. © 2018 American Society for Bone and Mineral Research.


Asunto(s)
Ameloblastos/citología , Ameloblastos/metabolismo , Diferenciación Celular , Odontoblastos/citología , Odontoblastos/metabolismo , Factor de Transcripción Sp7/metabolismo , Animales , Animales Recién Nacidos , Calcificación Fisiológica , Proliferación Celular , Colágeno/metabolismo , Dentina/metabolismo , Desarrollo Embrionario , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica , Incisivo/crecimiento & desarrollo , Incisivo/metabolismo , Incisivo/ultraestructura , Mesodermo/metabolismo , Ratones Endogámicos C57BL , Morfogénesis , Hueso Paladar/metabolismo , Transducción de Señal , Factor de Transcripción Sp7/deficiencia , Factor de Transcripción Sp7/genética , Células Madre/metabolismo
11.
Genesis ; 55(10)2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28921880

RESUMEN

Aggrecan (Acan), a large proteoglycan is abundantly expressed in cartilage tissue. Disruption of Acan gene causes dwarfism and perinatal lethality of homozygous mice. Because of sustained expression of Acan in the growth plate and articular cartilage, AgcCre model has been developed for the regulated ablation of target gene in chondrocytes. In this model, the IRES-CreERT-Neo-pgk transgene is knocked-in the 3'UTR of the Acan gene. We consistently noticed variable weight and size among the AgcCre littermates, prompting us to examine the cause of this phenotype. Wild-type, Cre-heterozygous (Agc+/Cre ), and Cre-homozygous (AgcCre/Cre ) littermates were indistinguishable at birth. However, by 1-month, AgcCre/Cre mice showed a significant reduction in body weight (18-27%) and body length (19-22%). Low body weight and dwarfism was sustained through adulthood and occurred in both genders. Compared with wild-type and Agc+/Cre littermates, long bones and vertebrae were shorter in AgcCre/Cre mice. Histological analysis of AgcCre/Cre mice revealed a significant reduction in the length of the growth plate and the thickness of articular cartilage. The amount of proteoglycan deposited in the cartilage of AgcCre/Cre mice was nearly half of the WT littermates. Analysis of gene expression indicates impaired differentiation of chondrocyte in hyaline cartilage of AgcCre/Cre mice. Notably, both Acan mRNA and protein was reduced by 50% in AgcCre/Cre mice. A strong correlation was noted between the level of Acan mRNA and the body length. Importantly, Agc+/Cre mice showed no overt skeletal phenotype. Thus to avoid misinterpretation of data, only the Agc+/Cre mice should be used for conditional deletion of a target gene in the cartilage tissue.


Asunto(s)
Agrecanos/genética , Enanismo/genética , Homocigoto , Agrecanos/metabolismo , Animales , Condrocitos/citología , Condrocitos/metabolismo , Enanismo/metabolismo , Enanismo/patología , Femenino , Regulación del Desarrollo de la Expresión Génica , Cartílago Hialino/metabolismo , Integrasas/genética , Integrasas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Osteogénesis , ARN Mensajero/genética , ARN Mensajero/metabolismo
12.
PLoS One ; 11(2): e0149119, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26901859

RESUMEN

RUNX1 a member of the family of runt related transcription factors (RUNX), is essential for hematopoiesis. The expression of RUNX1 gene is controlled by two promoters; the distal P1 promoter and the proximal P2 promoter. Several isoforms of RUNX1 mRNA are generated through the use of both promoters and alternative splicing. These isoforms not only differs in their temporal expression pattern but also exhibit differences in tissue specificity. The RUNX1 isoforms derived from P2 are expressed in a variety of tissues, but expression of P1-derived isoform is restricted to cells of hematopoietic lineage. However, the control of hematopoietic-cell specific expression is poorly understood. Here we report regulation of P1-derived RUNX1 mRNA by RUNX1 protein. In silico analysis of P1 promoter revealed presence of two evolutionary conserved RUNX motifs, 0.6kb upstream of the transcription start site, and three RUNX motifs within 170bp of the 5'UTR. Transcriptional contribution of these RUNX motifs was studied in myeloid and T-cells. RUNX1 genomic fragment containing all sites show very low basal activity in both cell types. Mutation or deletion of RUNX motifs in the UTR enhances basal activity of the RUNX1 promoter. Chromatin immunoprecipitation revealed that RUNX1 protein is recruited to these sites. Overexpression of RUNX1 in non-hematopoietic cells results in a dose dependent activation of the RUNX1 P1 promoter. We also demonstrate that RUNX1 protein regulates transcription of endogenous RUNX1 mRNA in T-cell. Finally we show that SCL transcription factor is recruited to regions containing RUNX motifs in the promoter and the UTR and regulates activity of the RUNX1 P1 promoter in vitro. Thus, multiple lines of evidence show that RUNX1 protein regulates its own gene transcription.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Regulación de la Expresión Génica , Regiones Promotoras Genéticas , Transcripción Genética , Regiones no Traducidas 5' , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Sitios de Unión , Línea Celular Tumoral , Humanos , Datos de Secuencia Molecular , Mutación , Motivos de Nucleótidos , Unión Proteica , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero , Alineación de Secuencia , Proteína 1 de la Leucemia Linfocítica T Aguda , Activación Transcripcional
13.
Oncotarget ; 7(10): 11299-309, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26849235

RESUMEN

Bone dissemination and bone disease occur in approximately 80% of patients with multiple myeloma (MM) and are a major cause of patient mortality. We previously demonstrated that MM cell-derived heparanase (HPSE) is a major driver of MM dissemination to and progression in new bone sites. However the mechanism(s) by which HPSE promotes MM progression remains unclear. In the present study, we investigated the involvement of mesenchymal features in HPSE-promoted MM progression in bone. Using a combination of molecular, biochemical, cellular, and in vivo approaches, we demonstrated that (1) HPSE enhanced the expression of mesenchymal markers in both MM and vascular endothelial cells; (2) HPSE expression in patient myeloma cells positively correlated with the expression of the mesenchymal markers vimentin and fibronectin. Additional mechanistic studies revealed that the enhanced mesenchymal-like phenotype induced by HPSE in MM cells is due, at least in part, to the stimulation of the ERK signaling pathway. Finally, knockdown of vimentin in HPSE expressing MM cells resulted in significantly attenuated MM cell dissemination and tumor growth in vivo. Collectively, these data demonstrate that the mesenchymal features induced by HPSE in MM cells contribute to enhanced tumor cell motility and bone-dissemination.


Asunto(s)
Movimiento Celular/fisiología , Glucuronidasa/metabolismo , Células Madre Mesenquimatosas/enzimología , Células Madre Mesenquimatosas/patología , Mieloma Múltiple/enzimología , Mieloma Múltiple/patología , Animales , Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Procesos de Crecimiento Celular/fisiología , Progresión de la Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Células Endoteliales/patología , Glucuronidasa/antagonistas & inhibidores , Heparina/análogos & derivados , Heparina/farmacología , Xenoinjertos , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones SCID , Mieloma Múltiple/metabolismo , Fenotipo , Transducción de Señal , Microambiente Tumoral , Vimentina/deficiencia , Vimentina/genética , Vimentina/metabolismo
14.
Mol Cell Biol ; 35(18): 3116-30, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26124283

RESUMEN

Studies of proteins involved in microRNA (miRNA) processing, maturation, and silencing have indicated the importance of miRNAs in skeletogenesis, but the specific miRNAs involved in this process are incompletely defined. Here, we identified miRNA 665 (miR-665) as a potential repressor of odontoblast maturation. Studies with cultured cell lines and primary embryonic cells showed that miR-665 represses the expression of early and late odontoblast marker genes and stage-specific proteases involved in dentin maturation. Notably, miR-665 directly targeted Dlx3 mRNA and decreased Dlx3 expression. Furthermore, RNA-induced silencing complex (RISC) immunoprecipitation and biotin-labeled miR-665 pulldown studies identified Kat6a as another potential target of miR-665. KAT6A interacted physically and functionally with RUNX2, activating tissue-specific promoter activity and prompting odontoblast differentiation. Overexpression of miR-665 reduced the recruitment of KAT6A to Dspp and Dmp1 promoters and prevented KAT6A-induced chromatin remodeling, repressing gene transcription. Taken together, our results provide novel molecular evidence that miR-665 functions in an miRNA-epigenetic regulatory network to control dentinogenesis.


Asunto(s)
Dentina/embriología , Dentinogénesis/genética , Regulación del Desarrollo de la Expresión Génica/genética , MicroARNs/genética , Odontoblastos/citología , Animales , Diferenciación Celular/genética , Línea Celular , Cromatina/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Silenciador del Gen , Células HEK293 , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Proteínas de Homeodominio/genética , Humanos , Ratones , MicroARNs/biosíntesis , Péptido Hidrolasas/biosíntesis , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Regiones Promotoras Genéticas/genética , Ratas , Sialoglicoproteínas/metabolismo , Factores de Transcripción/genética
15.
Blood ; 125(23): 3598-608, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-25862559

RESUMEN

The progression of multiple myeloma (MM) is governed by a network of molecular signals, the majority of which remain to be identified. Recent studies suggest that Runt-related transcription factor 2 (Runx2), a well-known bone-specific transcription factor, is also expressed in solid tumors, where expression promotes both bone metastasis and osteolysis. However, the function of Runx2 in MM remains unknown. The current study demonstrated that (1) Runx2 expression in primary human MM cells is significantly greater than in plasma cells from healthy donors and patients with monoclonal gammopathy of undetermined significance; (2) high levels of Runx2 expression in MM cells are associated with a high-risk population of MM patients; and (3) overexpression of Runx2 in MM cells enhanced tumor growth and disease progression in vivo. Additional studies demonstrated that MM cell-derived Runx2 promotes tumor progression through a mechanism involving the upregulation of Akt/ß-catenin/Survivin signaling and enhanced expression of multiple metastatic genes/proteins, as well as the induction of a bone-resident cell-like phenotype in MM cells. Thus, Runx2 expression supports the aggressive phenotype of MM and is correlated with poor prognosis. These data implicate Runx2 expression as a major regulator of MM progression in bone and myeloma bone disease.


Asunto(s)
Neoplasias Óseas/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/biosíntesis , Regulación Neoplásica de la Expresión Génica , Mieloma Múltiple/metabolismo , Osteólisis/metabolismo , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Línea Celular Tumoral , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Ratones , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Metástasis de la Neoplasia , Osteólisis/etiología , Osteólisis/genética , Osteólisis/patología , Proteínas Proto-Oncogénicas c-akt , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Survivin , beta Catenina/genética , beta Catenina/metabolismo
16.
J Bone Miner Res ; 30(1): 71-82, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25079226

RESUMEN

The Runx2 transcription factor is critical for commitment to the osteoblast lineage. However, its role in committed osteoblasts and its functions during postnatal skeletogenesis remain unclear. We established a Runx2-floxed line with insertion of loxP sites around exon 8 of the Runx2 gene. The Runx2 protein lacking the region encoded by exon 8 is imported into the nucleus and binds target DNA but exhibits diminished transcriptional activity. We specifically deleted the Runx2 gene in committed osteoblasts using 2.3-kb col1a-Cre transgenic mice. Surprisingly, the homozygous Runx2 mutant mice were born alive. The Runx2 heterozygous and homozygous null were grossly indistinguishable from wild-type littermates at birth. Runx2 deficiency did not alter proliferative capacity of osteoblasts during embryonic development (E18). Chondrocyte differentiation and cartilage growth in mutants was similar to wild-type mice from birth to 3 months of age. Analysis of the embryonic skeleton revealed poor calcification in homozygous mutants, which was more evident in bones formed by intramembranous ossification. Runx2 mutants showed progressive retardation in postnatal growth and exhibited significantly low bone mass by 1 month of age. Decreased bone formation was associated with decreased gene expression of osteoblast markers and impaired collagen assembly in the extracellular matrix. Consequently, Runx2 mutant bones exhibited decreased stiffness and structural integrity. By 3 months of age, bone acquisition in mutant mice was roughly half that of wild-type littermates. In addition to impaired osteoblast function, mutant mice showed markedly decreased osteoclast number and postnatal bone resorption. Taken together, functional deficiency of Runx2 in osteoblasts does not result in failed embryonic skeletogenesis but disrupts postnatal bone formation.


Asunto(s)
Desarrollo Óseo , Resorción Ósea/metabolismo , Diferenciación Celular , Proliferación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/deficiencia , Osteoblastos/metabolismo , Animales , Resorción Ósea/genética , Resorción Ósea/patología , Regulación de la Expresión Génica/genética , Homocigoto , Ratones , Ratones Noqueados , Osteoblastos/patología
17.
Connect Tissue Res ; 55 Suppl 1: 83-7, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25158187

RESUMEN

Runx2 and Sp7 transcription factors are essential for skeletogenesis. Targeted deletion of either gene results in failure of osteoblast differentiation and bone formation. Loss of bone-matrix gene expression is surprisingly similar in Sp7 and Runx2 null mice. The molecular mechanisms responsible for similar transcriptional regulation of target genes remain largely unknown. Here, we demonstrate that Runx2 and Sp7 interact physically and functionally. Both proteins are co-expressed in osteoblastic cells. We first characterized a panel of Sp7 antibodies and demonstrate that majority of the published antibodies do not recognize Sp7 protein. Co-immunoprecipitation studies revealed that endogenous Runx2 protein physically interacts with Sp7 protein. We identified that runt homology domain (RHD) of Runx2 protein is involved in physical association with Sp7. Functional consequences of Runx2-Sp7 physical interaction was then assessed by promoter-reporter assays. We selected promoters of osteocalcin (OC), a marker of mature osteoblast and fibroblast growth factor 3 (FGF3), a signaling molecule that determine the fate of embryonic ecto-mesenchyme. Runx2 and Sp7 stimulate OC-promoter activity by 3-folds in epithelial cells. However, when both proteins were co-expressed, a dose-dependent synergistic activation of 22-folds was noted. Similar pattern of synergistic activation of OC-promoter was noted in mesenchymal cell. FGF3 promoter was activated by 25 - and 30-folds with Runx2 and Sp7 respectively. Again a dose-dependent synergistic activation of 130-folds was evident when Runx2 and Sp7 were co-expressed in epithelial cells. Synergistic activation of FGF3 promoter was also noted in mesenchymal cells. Together, our data demonstrated that Runx2-Sp7 molecular complex functionally cooperate for maximal induction of cell-phenotype-restricted genes.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Regulación de la Expresión Génica , Osteoblastos/citología , Osteocalcina/metabolismo , Osteogénesis/fisiología , Factores de Transcripción/metabolismo , Diferenciación Celular/fisiología , Línea Celular , Regulación de la Expresión Génica/fisiología , Humanos , Unión Proteica , Factor de Transcripción Sp7
18.
Connect Tissue Res ; 55 Suppl 1: 88-91, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25158188

RESUMEN

Tooth formation is a multifaceted process involving numerous interactions between oral epithelium and neural crest derived ecto-mesenchyme from morphogenesis to cyto-differentiation. The precise molecular regulator that drives the cyto-differentiation and dynamic cross-talk between the two cell types has yet to be fully understood. Runx2 along with its downstream target Sp7 are essential transcription factors for development of the mineralizing cell types. Global knockout of the Runx2 gene results in an arrest of tooth morphogenesis at the late bud stage. Like Runx2, Sp7-null mutants exhibit peri-natal lethality and are completely devoid of alveolar bone. However, the role of Sp7 in tooth development remains elusive. Here, we report the effects of Sp7 deletion on tooth formation. Surprisingly, tooth morphogenesis progresses normally until the mid bell stage in Sp7-homozygous mutants. Incisors and multi-cusped first and second molars were noted in both littermates. Thus, formation of alveolar bone is not a prerequisite for tooth morphogenesis. Tooth organs of Sp7-null however, were significantly smaller in size when compared to WT. Differentiation of both ameloblasts and odontoblasts was disrupted in Sp7-null mice. Only premature and disorganized ameloblasts and odontoblasts were noted in mutant mice. These data indicate that Sp7 is not required for tooth morphogenesis but is obligatory for the functional maturation of both ameloblasts and odontoblasts.


Asunto(s)
Morfogénesis/genética , Odontoblastos/citología , Odontogénesis/genética , Diente/metabolismo , Factores de Transcripción/genética , Animales , Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Mesodermo/citología , Ratones Endogámicos C57BL , Ratones Noqueados , Diente Molar/crecimiento & desarrollo , Morfogénesis/fisiología , Factor de Transcripción Sp7 , Diente/embriología
19.
Connect Tissue Res ; 55 Suppl 1: 102-6, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25158191

RESUMEN

Runx2 transcription factor is essential for the development of mineralized tissue, and is required for osteoblast commitment and chondrocyte maturation. Mice with global deletion of Runx2 exhibit complete failure of bone tissue formation, while chondrocyte-specific Runx2-deficient mice lack endochondral ossification. However, the function of Runx2 after commitment of mesenchymal cells to the osteoblast lineage remains unknown. Here, we elucidate the osteoblast-specific requirements of Runx2 during development of the tissue. Runx2 was deleted in committed osteoblasts using Cre-recombinase driven by the 2.3kbCol1a1 promoter. Surprisingly, Runx2(ΔE8/ΔE8) mice were born alive and were essentially indistinguishable from wild-type littermates. At birth, we failed to detect any alterations in skeletal patterning or extent of bone development in homozygous mutants. However, by 4 weeks of age, mutant mice showed obvious growth deficiencies, and weighed 20-25% less than sex-matched wild-type littermates. Micro-CT analysis of the hindlimb revealed a dramatic decrease of 50% in both cortical and trabecular bone volume compared with wild-type mice. Consistent with this observation, trabecular number and thickness were decreased by 51% and 21%, respectively, and trabecular space was increased by 2-fold in limbs of Runx2(ΔE8/ΔE8) mice. In addition to poor acquisition of bone mass, the average density of hydroxyapatite was markedly decreased in bone of Runx2(ΔE8/ΔE8) mice. Together, these findings demonstrate that loss of Runx2 activity in committed osteoblasts impairs osteoblast function, and that Runx2 is critical for postnatal, but not embryonic endochondral ossification.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis/genética , Animales , Diferenciación Celular/genética , Células Madre Mesenquimatosas/citología , Ratones , Ratones Noqueados
20.
J Bone Miner Res ; 29(12): 2653-65, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24862038

RESUMEN

Synthesis of cartilage by chondrocytes is an obligatory step for endochondral ossification. Global deletion of the Runx2 gene results in complete failure of the ossification process, but the underlying cellular and molecular mechanisms are not fully known. Here, we elucidated Runx2 regulatory control distinctive to chondrocyte and cartilage tissue by generating Runx2 exon 8 floxed mice. Deletion of Runx2 gene in chondrocytes caused failure of endochondral ossification and lethality at birth. The limbs of Runx2(ΔE8/ΔE8) mice were devoid of mature chondrocytes, vasculature, and marrow. We demonstrate that the C-terminus of Runx2 drives its biological activity. Importantly, nuclear import and DNA binding functions of Runx2 are insufficient for chondrogenesis. Molecular studies revealed that despite normal levels of Sox9 and PTHrP, chondrocyte differentiation and cartilage growth are disrupted in Runx2(ΔE8/ΔE8) mice. Loss of Runx2 in chondrocytes also impaired osteoprotegerin-receptor activator of NF-κB ligand (OPG-RANKL) signaling and chondroclast development. Dwarfism observed in Runx2 mutants was associated with the near absence of proliferative zone in the growth plates. Finally, we show Runx2 directly regulates a unique set of cell cycle genes, Gpr132, Sfn, c-Myb, and Cyclin A1, to control proliferative capacity of chondrocyte. Thus, Runx2 is obligatory for both proliferation and differentiation of chondrocytes.


Asunto(s)
Diferenciación Celular/fisiología , Núcleo Celular/metabolismo , Proliferación Celular/fisiología , Condrocitos/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Osteogénesis/fisiología , Columna Vertebral/embriología , Transporte Activo de Núcleo Celular/fisiología , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/genética , Condrocitos/citología , Condrogénesis/fisiología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Ratones , Ratones Transgénicos , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Ligando RANK/genética , Ligando RANK/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Transducción de Señal/fisiología , Columna Vertebral/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...