Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Development ; 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39012257

RESUMEN

The Forkhead box transcription factors Foxc1 and Foxc2 are expressed in condensing mesenchyme cells at the onset of endochondral ossification. We used the Prx1-cre mouse to ablate Foxc1 and Foxc2 in limb skeletal progenitor cells. Prx1-cre;Foxc1Δ/ Δ;Foxc2Δ/Δ limbs were shorter than controls, with worsening phenotypes in distal structures. Cartilage formation and mineralization was severely disrupted in the paws. The radius and tibia were malformed, while the fibula and ulna remained unmineralized. Chondrocyte maturation was delayed with fewer Indian Hedgehog-expressing, prehypertrophic chondrocytes forming and a smaller hypertrophic chondrocyte zone. Later, progression out of chondrocyte hypertrophy was slowed, leading to an accumulation of COLX-expressing hypertrophic chondrocyte zone and formation of a smaller primary ossification center with fewer osteoblast progenitor cells populating this region. Targeting Foxc1 and Foxc2 in hypertrophic chondrocytes with Col10a1-cre also resulted in an expanded hypertrophic chondrocyte zone and smaller primary ossification center. Our findings suggest that Foxc1 and Foxc2 direct chondrocyte maturation towards hypertrophic chondrocyte formation. At later stages, Foxc1 and Foxc2 regulate function in hypertrophic chondrocyte remodelling to allow primary ossification center formation and osteoblast recruitment.

2.
bioRxiv ; 2023 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-37162896

RESUMEN

The forkhead box transcription factor genes Foxc1 and Foxc2 are expressed in the condensing mesenchyme of the developing skeleton prior to the onset of chondrocyte differentiation. To determine the roles of these transcription factors in limb development we deleted both Foxc1 and Foxc2 in lateral plate mesoderm using the Prx1-cre mouse line. Resulting compound homozygous mice died shortly after birth with exencephaly, and malformations to this sternum and limb skeleton. Notably distal limb structures were preferentially affected, with the autopods displaying reduced or absent mineralization. The radius and tibia bowed and the ulna and fibula were reduced to an unmineralized rudimentary structure. Molecular analysis revealed reduced expression of Ihh leading to reduced proliferation and delayed chondrocyte hypertrophy at E14.5. At later ages, Prx1-cre;Foxc1Δ/ Δ;Foxc2 Δ / Δ embryos exhibited restored Ihh expression and an expanded COLX-positive hypertrophic chondrocyte region, indicating a delayed exit and impaired remodeling of the hypertrophic chondrocytes. Osteoblast differentiation and mineralization were disrupted at the osteochondral junction and in the primary ossification center (POC). Levels of OSTEOPONTIN were elevated in the POC of compound homozygous mutants, while expression of Phex was reduced, indicating that impaired OPN processing by PHEX may underlie the mineralization defect we observe. Together our findings suggest that Foxc1 and Foxc2 act at different stages of endochondral ossification. Initially these genes act during the onset of chondrogenesis leading to the formation of hypertrophic chondrocytes. At later stages Foxc1 and Foxc2 are required for remodeling of HC and for Phex expression required for mineralization of the POC.

3.
Nat Commun ; 13(1): 7295, 2022 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-36435829

RESUMEN

While prior work has established that articular cartilage arises from Prg4-expressing perichondrial cells, it is not clear how this process is specifically restricted to the perichondrium of synovial joints. We document that the transcription factor Creb5 is necessary to initiate the expression of signaling molecules that both direct the formation of synovial joints and guide perichondrial tissue to form articular cartilage instead of bone. Creb5 promotes the generation of articular chondrocytes from perichondrial precursors in part by inducing expression of signaling molecules that block a Wnt5a autoregulatory loop in the perichondrium. Postnatal deletion of Creb5 in the articular cartilage leads to loss of both flat superficial zone articular chondrocytes coupled with a loss of both Prg4 and Wif1 expression in the articular cartilage; and a non-cell autonomous up-regulation of Ctgf. Our findings indicate that Creb5 promotes joint formation and the subsequent development of articular chondrocytes by driving the expression of signaling molecules that both specify the joint interzone and simultaneously inhibit a Wnt5a positive-feedback loop in the perichondrium.


Asunto(s)
Cartílago Articular , Fenómenos Fisiológicos Musculoesqueléticos , Cartílago Articular/metabolismo , Proteoglicanos/metabolismo , Condrocitos/metabolismo , Regulación de la Expresión Génica
4.
Bone ; 160: 116418, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35398294

RESUMEN

We previously found that FoxA factors are necessary for chondrocyte differentiation. To investigate whether FoxA factors alone are sufficient to drive chondrocyte hypertrophy, we build a FoxA2 transgenic mouse in which FoxA2 cDNA is driven by a reiterated Tetracycline Response Element (TRE) and a minimal CMV promoter. This transgenic line was crossed with a col2CRE;Rosa26rtTA/+ mouse line to generate col2CRE;Rosa26rtTA/+;TgFoxA2+/- mice for inducible expression of FoxA2 in cartilage using doxycycline treatment. Ectopic expression of FoxA2 in the developing skeleton reveals skeletal defects and shorter skeletal elements in E17.5 mice. The chondro-osseous border was frequently mis-shaped in mutant mice, with small islands of col.10+ hypertrophic cells extending in the metaphyseal bone. Even though overexpression of FoxA2 causes an accumulation of hypertrophic chondrocytes, it did not trigger ectopic hypertrophy in the immature chondrocytes. This suggests that FoxA2 may need transcriptional co-factors (such as Runx2), whose expression is restricted to the hypertrophic zone, and absent in the immature chondrocytes. To investigate a potential FoxA2/Runx2 interaction in immature chondrocytes versus hypertrophic cells, we separated these two subpopulations by FACS to obtain CD24+CD200+ hypertrophic chondrocytes and CD24+CD200- immature chondrocytes and we ectopically expressed FoxA2 alone or in combination with Runx2 via lentiviral gene delivery. In CD24+CD200+ hypertrophic chondrocytes, FoxA2 enhanced the expression of chondrocyte hypertrophic markers collagen 10, MMP13, and alkaline phosphatase. In contrast, in the CD24+CD200- immature chondrocytes, neither FoxA2 nor Runx2 overexpression could induce ectopic expression of hypertrophic markers MMP13, alkaline phosphatase, or PTH/PTHrP receptor. Overall these findings mirror our in vivo data, and suggest that induction of chondrocyte hypertrophy by FoxA2 may require other factors in addition to Runx2 (i.e., Hif2α, MEF2C, or perhaps unknown factors), whose expression/activity is rate-limiting in immature chondrocytes.


Asunto(s)
Condrocitos , Subunidad alfa 1 del Factor de Unión al Sitio Principal , Fosfatasa Alcalina/metabolismo , Animales , Huesos/metabolismo , Cartílago/metabolismo , Diferenciación Celular/genética , Condrocitos/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/metabolismo , Hipertrofia , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Ratones , Factores de Transcripción/metabolismo
5.
Commun Biol ; 4(1): 332, 2021 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-33712729

RESUMEN

A hallmark of cells comprising the superficial zone of articular cartilage is their expression of lubricin, encoded by the Prg4 gene, that lubricates the joint and protects against the development of arthritis. Here, we identify Creb5 as a transcription factor that is specifically expressed in superficial zone articular chondrocytes and is required for TGF-ß and EGFR signaling to induce Prg4 expression. Notably, forced expression of Creb5 in chondrocytes derived from the deep zone of the articular cartilage confers the competence for TGF-ß and EGFR signals to induce Prg4 expression. Chromatin-IP and ATAC-Seq analyses have revealed that Creb5 directly binds to two Prg4 promoter-proximal regulatory elements, that display an open chromatin conformation specifically in superficial zone articular chondrocytes; and which work in combination with a more distal regulatory element to drive induction of Prg4 by TGF-ß. Our results indicate that Creb5 is a critical regulator of Prg4/lubricin expression in the articular cartilage.


Asunto(s)
Cartílago Articular/metabolismo , Condrocitos/metabolismo , Proteína de Unión al Elemento de Respuesta al AMP Cíclico/metabolismo , Proteoglicanos/metabolismo , Animales , Sitios de Unión , Cartílago Articular/efectos de los fármacos , Bovinos , Células Cultivadas , Condrocitos/efectos de los fármacos , Proteína de Unión al Elemento de Respuesta al AMP Cíclico/genética , Regulación de la Expresión Génica , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Proteoglicanos/genética , Factor de Crecimiento Transformador alfa/farmacología , Factor de Crecimiento Transformador beta2/farmacología
6.
JCI Insight ; 4(5)2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30843886

RESUMEN

During endochondral bone formation, chondrocyte hypertrophy represents a crucial turning point from chondrocyte differentiation to bone formation. Both parathyroid hormone-related protein (PTHrP) and histone deacetylase 4 (HDAC4) inhibit chondrocyte hypertrophy. Using multiple mouse genetics models, we demonstrate in vivo that HDAC4 is required for the effects of PTHrP on chondrocyte differentiation. We further show in vivo that PTHrP leads to reduced HDAC4 phosphorylation at the 14-3-3-binding sites and subsequent HDAC4 nuclear translocation. The Hdac4-KO mouse shares a similar but milder phenotype with the Pthrp-KO mouse, indicating the possible existence of other mediators of PTHrP action. We identify HDAC5 as an additional mediator of PTHrP signaling. While the Hdac5-KO mouse has no growth plate phenotype at birth, the KO of Hdac5 in addition to the KO of Hdac4 is required to block fully PTHrP action on chondrocyte differentiation at birth in vivo. Finally, we show that PTHrP suppresses myocyte enhancer factor 2 (Mef2) action that allows runt-related transcription factor 2 (Runx2) mRNA expression needed for chondrocyte hypertrophy. Our results demonstrate that PTHrP inhibits chondrocyte hypertrophy and subsequent bone formation in vivo by allowing HDAC4 and HDAC5 to block the Mef2/Runx2 signaling cascade. These results explain the phenotypes of several genetic abnormalities in humans.


Asunto(s)
Condrocitos/metabolismo , Histona Desacetilasas/metabolismo , Hipertrofia/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Animales , Cartílago/patología , Proliferación Celular , Condrocitos/patología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Histona Desacetilasas/genética , Humanos , Hipertrofia/genética , Factores de Transcripción MEF2/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteogénesis/genética , Osteogénesis/fisiología , Proteína Relacionada con la Hormona Paratiroidea/genética , Fenotipo , Fosforilación , ARN Mensajero/metabolismo , Costillas/patología , Transducción de Señal , Transcriptoma
7.
Semin Cell Dev Biol ; 72: 3-9, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29097153

RESUMEN

In 1987, Robert Davis, Hal Weintraub and I reported the identification of MyoD, a transcription factor that could reprogram fibroblasts into skeletal muscle cells. In this recollection, I both summarize the prior work of Helen Blau, Woody Wright, Peter Jones and Charlie Emerson that inspired my entry into this field, and the subsequent events that led to finding MyoD. Lastly, I highlight some of the principles in developmental biology that have emerged during the past 30 years, which are particularly relevant to skeletal muscle biology.


Asunto(s)
Fibroblastos/metabolismo , Desarrollo de Músculos/genética , Músculo Esquelético/metabolismo , Proteína MioD/genética , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Fibroblastos/citología , Regulación del Desarrollo de la Expresión Génica , Ratones , Músculo Esquelético/citología , Músculo Esquelético/embriología
8.
FASEB J ; 31(3): 1067-1084, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27965322

RESUMEN

Articular cartilage has little regenerative capacity. Recently, genetic lineage tracing experiments have revealed chondrocyte progenitors at the articular surface. We further characterized these progenitors by using in vivo genetic approaches. Histone H2B-green fluorescent protein retention revealed that superficial cells divide more slowly than underlying articular chondrocytes. Clonal genetic tracing combined with immunohistochemistry revealed that superficial cells renew their number by symmetric division, express mesenchymal stem cell markers, and generate chondrocytes via both asymmetric and symmetric differentiation. Quantitative analysis of cellular kinetics, in combination with phosphotungstic acid-enhanced micro-computed tomography, showed that superficial cells generate chondrocytes and contribute to the growth and reshaping of articular cartilage. Furthermore, we found that cartilage renewal occurs as the progeny of superficial cells fully replace fetal chondrocytes during early postnatal life. Thus, superficial cells are self-renewing progenitors that are capable of maintaining their own population and fulfilling criteria of unipotent adult stem cells. Furthermore, the progeny of these cells reconstitute adult articular cartilage de novo, entirely substituting fetal chondrocytes.-Li, L., Newton, P. T., Bouderlique, T., Sejnohova, M., Zikmund, T., Kozhemyakina, E., Xie, M., Krivanek, J., Kaiser, J., Qian, H., Dyachuk, V., Lassar, A. B., Warman, M. L., Barenius, B., Adameyko, I., Chagin, A. S. Superficial cells are self-renewing chondrocyte progenitors, which form the articular cartilage in juvenile mice.


Asunto(s)
Células Madre Adultas/citología , Cartílago Articular/citología , Condrocitos/citología , Condrogénesis , Animales , Cartílago Articular/fisiología , Ratones , Regeneración
9.
Development ; 142(5): 817-31, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25715393

RESUMEN

Decades of work have identified the signaling pathways that regulate the differentiation of chondrocytes during bone formation, from their initial induction from mesenchymal progenitor cells to their terminal maturation into hypertrophic chondrocytes. Here, we review how multiple signaling molecules, mechanical signals and morphological cell features are integrated to activate a set of key transcription factors that determine and regulate the genetic program that induces chondrogenesis and chondrocyte differentiation. Moreover, we describe recent findings regarding the roles of several signaling pathways in modulating the proliferation and maturation of chondrocytes in the growth plate, which is the 'engine' of bone elongation.


Asunto(s)
Condrocitos/citología , Condrocitos/metabolismo , Factores de Transcripción/metabolismo , Animales , Condrocitos/fisiología , Condrogénesis/genética , Condrogénesis/fisiología , Placa de Crecimiento/citología , Placa de Crecimiento/metabolismo , Placa de Crecimiento/fisiología , Humanos , Factores de Transcripción/genética
10.
Arthritis Rheumatol ; 67(5): 1261-73, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25603997

RESUMEN

OBJECTIVE: To generate knockin mice that express a tamoxifen-inducible Cre recombinase from the Prg4 locus (Prg4(GFPCreERt2) mice) and to use these animals to fate-map the progeny of Prg4-positive articular cartilage cells at various ages. METHODS: We crossed Prg4(GFPCreERt2) mice with Rosa26(floxlacZ) or Rosa26(mTmG) reporter strains, admin-istered tamoxifen to the double heterozygous offspring at different ages, and assayed Cre-mediated recom-bination by histochemistry and/or fluorescence microscopy. RESULTS: In 1-month-old mice, the expression of the Prg4(GFPCreERt2) allele mirrored the expression of endogenous Prg4 and, when tamoxifen was admin-istered for 10 days, caused Cre-mediated recombination in ∼70% of the superficial-most chondrocytes. Prg4(GFPCreERt2)-expressing cells were mostly confined to the top 3 cell layers of the articular cartilage in 1-month-old mice, but descendants of these cells were located in deeper regions of the articular cartilage in aged mice. On embryonic day 17.5, Prg4(GFPCreERt2)-expressing cells were largely restricted to the superficial-most cell layer of the forming joint, yet at ∼1 year, the progeny of these cells spanned the depth of the articular cartilage. CONCLUSION: Our results suggest that Prg4-expressing cells located at the joint surface in the embryo serve as a progenitor population for all deeper layers of the mature articular cartilage. Also, our findings indicate that Prg4(GFPCreERt2) is expressed by superficial chondrocytes in young mice, but expands into deeper regions of the articular cartilage as the animals age. The Prg4(GFPCreERt2) allele should be a useful tool for inducing efficient Cre-mediated recombination of loxP-flanked alleles at sites of Prg4 expression.


Asunto(s)
Cartílago Articular/metabolismo , Condrocitos/metabolismo , Proteoglicanos/metabolismo , Células Madre/metabolismo , Animales , Cartílago Articular/citología , Condrocitos/citología , Técnicas de Sustitución del Gen , Integrasas , Ratones , Proteoglicanos/genética , Células Madre/citología
11.
Development ; 141(20): 3978-87, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25294942

RESUMEN

The relative timing of SHH and BMP signals controls whether presomitic mesoderm (PSM) cells will adopt either a chondrogenic or lateral plate mesoderm fate. Here we document that SHH-mediated induction of Nkx3.2 maintains the competence of somitic cells to initiate chondrogenesis in response to subsequent BMP signals by repressing BMP-dependent induction of GATA genes. Conversely, administration of BMP signals to PSM or forced expression of GATA family members in chick PSM explants blocks induction of hedgehog-dependent gene expression. We demonstrate that GATA factors can interact with Gli factors and can recruit the transcriptional co-factor FOG1 (ZFPM1) to the regulatory region of the mouse Gli1 gene, repressing the induction of Gli1 by SHH by binding to both GATA and Gli binding sites. Knockdown of FOG1 reverses the ability of GATA factors to repress Gli1 expression. Our findings uncover a novel role for GATA transcription factors as repressors of hedgehog signaling, and document that NKX3.2 maintains the ability of sclerotomal cells to express SHH transcriptional targets in the presence of BMP signals by repressing the induction of Gata4/5/6.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Factor de Transcripción GATA4/metabolismo , Factor de Transcripción GATA5/metabolismo , Factor de Transcripción GATA6/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Animales , Condrocitos/citología , Perfilación de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Células 3T3 NIH , Proteínas Nucleares/metabolismo , Proteína con Dedos de Zinc GLI1
12.
Cell Rep ; 8(5): 1419-31, 2014 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-25159139

RESUMEN

The formation of cartilage is restricted to the core of the limb bud mesenchyme by ectodermal Wnts, which can irreversibly silence expression of the prochondrogenic transcription factor Sox9. In contrast, fibroblast growth factor (FGF) signals from the apical ectodermal ridge maintain the competence of chondrogenic precursors to undergo chondrogenesis once these cells go out of the range of ectodermal Wnt signals. We have found that Wnt signals induce both a repressive chromatin mark (H3K27me3) and DNA methylation over the Sox9 promoter and that Wnt-induced irreversible silencing of the Sox9 gene requires DNA methylation of this locus, which is specifically countered by FGF signals. FGF blocks the recruitment of the de novo DNA methyltransferase, DNMT3A, to the Sox9 promoter by inducing the interaction and phosphorylation of DNMT3A by ERK1/ERK2 and thereby controls whether expression of Sox9 is either irreversibly or reversibly silenced by Wnt signals in limb bud mesenchymal cells.


Asunto(s)
Condrogénesis , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Factores de Crecimiento de Fibroblastos/farmacología , Esbozos de los Miembros/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales , Células Cultivadas , Embrión de Pollo , Metilación de ADN , ADN Metiltransferasa 3A , Células HEK293 , Humanos , Esbozos de los Miembros/citología , Esbozos de los Miembros/embriología , Sistema de Señalización de MAP Quinasas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Vía de Señalización Wnt
13.
Genes Dev ; 28(2): 127-39, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24449269

RESUMEN

Lubricin is a secreted proteoglycan encoded by the Prg4 locus that is abundantly expressed by superficial zone articular chondrocytes and has been noted to both be sensitive to mechanical loading and protect against the development of osteoarthritis. In this study, we document that running induces maximal expression of Prg4 in the superficial zone of knee joint articular cartilage in a COX-2-dependent fashion, which correlates with augmented levels of phospho-S133 CREB and increased nuclear localization of CREB-regulated transcriptional coactivators (CRTCs) in this tissue. Furthermore, we found that fluid flow shear stress (FFSS) increases secretion of extracellular PGE2, PTHrP, and ATP (by epiphyseal chondrocytes), which together engage both PKA- and Ca(++)-regulated signaling pathways that work in combination to promote CREB-dependent induction of Prg4, specifically in superficial zone articular chondrocytes. Because running and FFSS both boost Prg4 expression in a COX-2-dependent fashion, our results suggest that mechanical motion may induce Prg4 expression in the superficial zone of articular cartilage by engaging the same signaling pathways activated in vitro by FFSS that promote CREB-dependent gene expression in this tissue.


Asunto(s)
Cartílago Articular/metabolismo , Regulación de la Expresión Génica , Proteoglicanos/genética , Proteoglicanos/metabolismo , Transducción de Señal , Adenosina Trifosfato/metabolismo , Alelos , Animales , Proteína de Unión a CREB/metabolismo , Calcio/metabolismo , Células Cultivadas , Condrocitos/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Masculino , Ratones , Actividad Motora/genética , Recombinación Genética/genética , Estrés Fisiológico/genética
14.
PLoS Genet ; 10(1): e1004072, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24415953

RESUMEN

In the limb bud, patterning along the anterior-posterior (A-P) axis is controlled by Sonic Hedgehog (Shh), a signaling molecule secreted by the "Zone of Polarizing Activity", an organizer tissue located in the posterior margin of the limb bud. We have found that the transcription factors GATA4 and GATA6, which are key regulators of cell identity, are expressed in an anterior to posterior gradient in the early limb bud, raising the possibility that GATA transcription factors may play an additional role in patterning this tissue. While both GATA4 and GATA6 are expressed in an A-P gradient in the forelimb buds, the hindlimb buds principally express GATA6 in an A-P gradient. Thus, to specifically examine the role of GATA6 in limb patterning we generated Prx1-Cre; GATA6(fl/fl) mice, which conditionally delete GATA6 from their developing limb buds. We found that these animals display ectopic expression of both Shh and its transcriptional targets specifically in the anterior mesenchyme of the hindlimb buds. Loss of GATA6 in the developing limbs results in the formation of preaxial polydactyly in the hindlimbs. Conversely, forced expression of GATA6 throughout the limb bud represses expression of Shh and results in hypomorphic limbs. We have found that GATA6 can bind to chromatin (isolated from limb buds) encoding either Shh or Gli1 regulatory elements that drive expression of these genes in this tissue, and demonstrated that GATA6 works synergistically with FOG co-factors to repress expression of luciferase reporters driven by these sequences. Most significantly, we have found that conditional loss of Shh in limb buds lacking GATA6 prevents development of hindlimb polydactyly in these compound mutant embryos, indicating that GATA6 expression in the anterior region of the limb bud blocks hindlimb polydactyly by repressing ectopic expression of Shh.


Asunto(s)
Tipificación del Cuerpo/genética , Factor de Transcripción GATA6/biosíntesis , Proteínas Hedgehog/metabolismo , Esbozos de los Miembros/metabolismo , Polidactilia/genética , Animales , Embrión de Mamíferos , Desarrollo Embrionario , Miembro Anterior/crecimiento & desarrollo , Miembro Anterior/metabolismo , Factor de Transcripción GATA4/biosíntesis , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/metabolismo , Factor de Transcripción GATA6/genética , Factor de Transcripción GATA6/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/biosíntesis , Proteínas Hedgehog/genética , Miembro Posterior/crecimiento & desarrollo , Miembro Posterior/metabolismo , Ratones , Polidactilia/etiología , Polidactilia/patología , Transducción de Señal/genética
15.
Dev Biol ; 374(1): 198-209, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23165293

RESUMEN

The cardiac homeobox gene Nkx2.5 plays a key and dosage-sensitive role in the differentiation of outflow tract and right ventricle from progenitors of the second heart field (SHF) and Nkx2.5 mutation is strongly associated with human outflow tract congenital heart disease (OFT CHD). Therefore defining the regulatory mechanisms controlling Nkx2.5 expression in SHF populations serves an important function in understanding the etiology of complex CHD. Through a comparative analysis of regulatory elements controlling SHF expression of Nkx2.5 in the chicken and mouse, we have found evidence that Nkx2.5 autoregulation is important for maintaining Nkx2.5 expression during SHF differentiation in both species. However the mechanism of Nkx2.5 maintenance differs between placental mammals and non-mammalian vertebrates: in chick Nkx2.5 binds directly to a genomic enhancer element that is required to maintain Nkx2.5 expression in the SHF. In addition, it is likely that this is true in other non-mammalian vertebrates given that they possess a similar genomic organization. By contrast, in placental mammals, Nkx2.5 autoregulation in the SHF functions indirectly through Mef2c. These data underscore a tight relationship in mammals between Nkx2.5 and Mef2c in SHF transcriptional regulation, and highlight the potential for evolutionary cis-regulatory analysis to identify core, conserved components of the gene networks controlling heart development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Miocardio/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Animales , Secuencia de Bases , Pollos , Elementos de Facilitación Genéticos , Perfilación de la Expresión Génica , Vectores Genéticos , Insuficiencia Cardíaca/congénito , Insuficiencia Cardíaca/metabolismo , Proteína Homeótica Nkx-2.5 , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Células Musculares/citología , Homología de Secuencia de Ácido Nucleico , Células Madre/citología
16.
Dev Cell ; 22(5): 927-39, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22595668

RESUMEN

During endochondral ossification, small, immature chondrocytes enlarge to form hypertrophic chondrocytes, which express collagen X. In this work, we demonstrate that FoxA factors are induced during chondrogenesis, bind to conserved binding sites in the collagen X enhancer, and can promote the expression of a collagen X-luciferase reporter in both chondrocytes and fibroblasts. In addition, we demonstrate by both gain- and loss-of-function analyses that FoxA factors play a crucial role in driving the expression of both endogenous collagen X and other hypertrophic chondrocyte-specific genes. Mice engineered to lack expression of both FoxA2 and FoxA3 in their chondrocytes display defects in chondrocyte hypertrophy, alkaline phosphatase expression, and mineralization in their sternebrae and, in addition, exhibit postnatal dwarfism that is coupled to significantly decreased expression of both collagen X and MMP13 in their growth plates. Our findings indicate that FoxA family members are crucial regulators of the hypertrophic chondrocyte differentiation program.


Asunto(s)
Aumento de la Célula , Condrocitos/metabolismo , Condrogénesis/genética , Colágeno Tipo X/metabolismo , Enanismo/genética , Factor Nuclear 3-beta del Hepatocito/metabolismo , Factor Nuclear 3-gamma del Hepatocito/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Sitios de Unión , Diferenciación Celular/genética , Células Cultivadas , Embrión de Pollo , Condrocitos/citología , Colágeno Tipo X/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Enanismo/embriología , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Genes Reporteros , Placa de Crecimiento/metabolismo , Factor Nuclear 3-beta del Hepatocito/deficiencia , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 3-gamma del Hepatocito/deficiencia , Factor Nuclear 3-gamma del Hepatocito/genética , Metaloproteinasa 13 de la Matriz/genética , Huesos Metatarsianos/citología , Huesos Metatarsianos/metabolismo , Ratones , Ratones Mutantes , Factores Reguladores Miogénicos/metabolismo , Proteína Smad1/metabolismo
17.
Dev Biol ; 353(1): 29-37, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21354132

RESUMEN

In the avian embryo, endothelial cells originate from several sources, including the lateral plate and somite mesoderm. In this study, we show that Gata transcription factors are expressed in the lateral plate and in vasculogenic regions of the avian somite and are able to promote a vascular endothelial fate when ectopically expressed in somite precursors. A fusion of GATA4 to the transcriptional activator VP16 promoted endothelium formation, indicating that GATA transcription factors promote vasculogenesis via activation of downstream targets, while a fusion of GATA4 to the transcriptional repressor engrailed repressed expression of Vascular Endothelial Growth Factor Receptor 2, a marker of endothelial precursors. These findings indicate a role for GATA transcription factors in the differentiation of the endothelium.


Asunto(s)
Proteínas Aviares/fisiología , Diferenciación Celular , Células Endoteliales/citología , Factores de Transcripción GATA/fisiología , Animales , Apoptosis , Proteína Morfogenética Ósea 2/farmacología , Embrión de Pollo , Coturnix/embriología , Mesodermo/patología
18.
J Cell Biol ; 187(7): 941-3, 2009 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-20026653

RESUMEN

In this issue, Gillespie et al. (Gillespie et al. 2009. J. Cell Biol. doi:10.1083/jcb.200907037) demonstrate that the mitogen-activated protein kinase isoform p38-gamma plays a crucial role in blocking the premature differentiation of satellite cells, a skeletal muscle stem cell population. p38-gamma puts the brakes on skeletal muscle differentiation by promoting the association of the transcription factor MyoD with the histone methyltransferase, KMT1A, which act together in a complex to repress the premature expression of the gene encoding the myogenic transcription factor Myogenin.


Asunto(s)
Diferenciación Celular/fisiología , Músculo Esquelético/citología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Animales , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , Ratones , Músculo Esquelético/metabolismo , Proteína MioD/metabolismo
19.
Mol Cell Biol ; 29(21): 5751-62, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19704004

RESUMEN

The maturation of immature chondrocytes to hypertrophic chondrocytes is regulated by parathyroid hormone-related peptide (PTHrP). We demonstrate that PTHrP or forskolin administration can block induction of collagen X-luciferase by exogenous Runx2, MEF2, and Smad1 in transfected chondrocytes. We have found that PTHrP/forskolin administration represses the transcriptional activity of MEF2 and that forced expression of MEF2-VP16 can restore expression of the collagen X reporter in chondrocytes treated with these agents. PTHrP/forskolin induces dephosphorylation of histone deacetylase 4 (HDAC4) phospho-S246, which decreases interaction of HDAC4 with cytoplasmic 14-3-3 proteins and promotes nuclear translocation of HDAC4 and repression of MEF2 transcriptional activity. We have found that forskolin increases the activity of an HDAC4 phospho-S246 phosphatase and that forskolin-induced nuclear translocation of HDAC4 was reversed by the protein phosphatase 2A (PP2A) antagonist, okadaic acid. Finally, we demonstrate that knockdown of PP2A inhibits forskolin-induced nuclear translocation of HDAC4 and attenuates the ability of this signaling molecule to repress collagen X expression in chondrocytes, indicating that PP2A is critical for PTHrP-mediated regulation of chondrocyte hypertrophy.


Asunto(s)
Condrocitos/enzimología , Condrocitos/patología , Histona Desacetilasas/metabolismo , Factores Reguladores Miogénicos/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/farmacología , Proteína Fosfatasa 2/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas 14-3-3/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Pollos , Condrocitos/efectos de los fármacos , Colforsina/farmacología , Colágeno Tipo X/genética , Colágeno Tipo X/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Activación Enzimática/efectos de los fármacos , Genes Reporteros , Hipertrofia , Luciferasas/metabolismo , Factores de Transcripción MEF2 , Ratones , Ácido Ocadaico/farmacología , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Proteínas Recombinantes de Fusión/metabolismo , Proteína Smad1/genética , Proteína Smad4/genética , Transcripción Genética/efectos de los fármacos
20.
Mol Cell Biol ; 29(15): 4262-73, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19470758

RESUMEN

In this study, we demonstrate that dedifferentiation of round primary chondrocytes into a fibroblast morphology correlates with a profound induction of RhoA protein and stress fibers. Culture of dedifferentiated chondrocytes in alginate gel induces a precipitous loss of RhoA protein and a loss of stress fibers concomitant with the reexpression of the chondrocyte differentiation program. We have found that chondrogenesis in limb bud micromass cultures similarly entails a loss of RhoA protein and that expression of dominant negative RhoA in such cultures can markedly enhance chondrogenesis. Consistent with these results, expression of the Rho antagonist C3 transferase can restore chondrocyte gene expression in dedifferentiated chondrocytes grown on plastic. Transfection of cells with agents that block actin polymerization enhance the ability of either exogenous Sox9 or a Gal4 DBD-Sox9 fusion protein to activate gene expression. Interestingly, the enhancement of Sox9 function by actin depolymerization requires both protein kinase A (PKA) activity and a PKA phosphorylation site in Sox9 (S181) that is known to enhance Sox9 transcriptional activity. Lastly, we demonstrate that RhoA-mediated modulation of actin polymerization regulates the ability of Sox9 to both activate chondrocyte-specific markers and maintain its own expression in chondrocytes via a positive feedback loop.


Asunto(s)
Actinas/metabolismo , Factor de Transcripción SOX9/genética , Transducción de Señal , Transcripción Genética/genética , Proteína de Unión al GTP rhoA/metabolismo , Animales , Sitios de Unión/genética , Western Blotting , Desdiferenciación Celular , Células Cultivadas , Embrión de Pollo , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Técnica del Anticuerpo Fluorescente , Isoquinolinas/farmacología , Mutación , Fosforilación , Polímeros/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Multimerización de Proteína , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción SOX9/metabolismo , Sulfonamidas/farmacología , Proteína de Unión al GTP rhoA/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...