Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34161280

RESUMO

Osteogenesis imperfecta (OI) is a genetic disorder that features wide-ranging defects in both skeletal and nonskeletal tissues. Previously, we and others reported that loss-of-function mutations in FK506 Binding Protein 10 (FKBP10) lead to skeletal deformities in conjunction with joint contractures. However, the pathogenic mechanisms underlying joint dysfunction in OI are poorly understood. In this study, we have generated a mouse model in which Fkbp10 is conditionally deleted in tendons and ligaments. Fkbp10 removal substantially reduced telopeptide lysyl hydroxylation of type I procollagen and collagen cross-linking in tendons. These biochemical alterations resulting from Fkbp10 ablation were associated with a site-specific induction of fibrosis, inflammation, and ectopic chondrogenesis followed by joint deformities in postnatal mice. We found that the ectopic chondrogenesis coincided with enhanced Gli1 expression, indicating dysregulated Hedgehog (Hh) signaling. Importantly, genetic inhibition of the Hh pathway attenuated ectopic chondrogenesis and joint deformities in Fkbp10 mutants. Furthermore, Hh inhibition restored alterations in gait parameters caused by Fkbp10 loss. Taken together, we identified a previously unappreciated role of Fkbp10 in tendons and ligaments and pathogenic mechanisms driving OI joint dysfunction.


Assuntos
Condrócitos/patologia , Articulações/fisiopatologia , Atividade Motora , Osteogênese Imperfeita/fisiopatologia , Osteogênese , Proteínas de Ligação a Tacrolimo/metabolismo , Animais , Animais Recém-Nascidos , Condrogênese/genética , Colágeno Tipo I/metabolismo , Modelos Animais de Doenças , Fibrose , Marcha , Deleção de Genes , Regulação da Expressão Gênica , Proteínas Hedgehog/metabolismo , Hidroxilação , Inflamação/genética , Inflamação/patologia , Articulações/patologia , Ligamentos/patologia , Lisina/metabolismo , Camundongos , Modelos Biológicos , Ossificação Heterotópica/complicações , Ossificação Heterotópica/genética , Ossificação Heterotópica/patologia , Ossificação Heterotópica/fisiopatologia , Osteogênese/genética , Osteogênese Imperfeita/complicações , Osteogênese Imperfeita/genética , Osteogênese Imperfeita/patologia , Peptídeos/metabolismo , Análise de Sequência de RNA , Transdução de Sinais , Proteínas de Ligação a Tacrolimo/genética , Tendões/patologia
2.
PLoS Biol ; 18(11): e3000902, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33201874

RESUMO

Coordinated development of muscles, tendons, and their attachment sites ensures emergence of functional musculoskeletal units that are adapted to diverse anatomical demands among different species. How these different tissues are patterned and functionally assembled during embryogenesis is poorly understood. Here, we investigated the morphogenesis of extraocular muscles (EOMs), an evolutionary conserved cranial muscle group that is crucial for the coordinated movement of the eyeballs and for visual acuity. By means of lineage analysis, we redefined the cellular origins of periocular connective tissues interacting with the EOMs, which do not arise exclusively from neural crest mesenchyme as previously thought. Using 3D imaging approaches, we established an integrative blueprint for the EOM functional unit. By doing so, we identified a developmental time window in which individual EOMs emerge from a unique muscle anlage and establish insertions in the sclera, which sets these muscles apart from classical muscle-to-bone type of insertions. Further, we demonstrate that the eyeballs are a source of diffusible all-trans retinoic acid (ATRA) that allow their targeting by the EOMs in a temporal and dose-dependent manner. Using genetically modified mice and inhibitor treatments, we find that endogenous local variations in the concentration of retinoids contribute to the establishment of tendon condensations and attachment sites that precede the initiation of muscle patterning. Collectively, our results highlight how global and site-specific programs are deployed for the assembly of muscle functional units with precise definition of muscle shapes and topographical wiring of their tendon attachments.


Assuntos
Músculos Oculomotores/embriologia , Músculos Oculomotores/crescimento & desenvolvimento , Tretinoína/metabolismo , Animais , Tecido Conjuntivo/fisiologia , Desenvolvimento Embrionário , Olho , Imageamento Tridimensional/métodos , Camundongos/embriologia , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Morfogênese , Transdução de Sinais , Tendões/fisiologia , Tretinoína/fisiologia
3.
Dev Biol ; 470: 108-120, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33248111

RESUMO

Growth of the musculoskeletal system requires precise coordination between bone, muscle, and tendon during development. Insufficient elongation of the muscle-tendon unit relative to bone growth results in joint contracture, a condition characterized by reduction or complete loss of joint range of motion. Here we establish a novel murine model of joint contracture by targeting Smad4 for deletion in the tendon cell lineage using Scleraxis-Cre (ScxCre). Smad4ScxCre mutants develop a joint contracture shortly after birth. The contracture is stochastic in direction and increases in severity with age. Smad4ScxCre mutant tendons exhibited a stable reduction in cellularity and a progressive reduction in extracellular matrix volume. Collagen fibril diameters were reduced in the Smad4ScxCre mutants, suggesting a role for Smad4 signaling in the regulation of matrix accumulation. Although ScxCre also has sporadic activity in both cartilage and muscle, we demonstrate an essential role for Smad4 loss in tendons for the development of joint contractures. Disrupting the canonical TGFß-pathway in Smad2;3ScxCre mutants did not result in joint contractures. Conversely, disrupting the BMP pathway by targeting BMP receptors (Alk3ScxCre/Alk6null) recapitulated many features of the Smad4ScxCre contracture phenotype, suggesting that joint contracture in Smad4ScxCre mutants is caused by disruption of BMP signaling. Overall, these results establish a model of murine postnatal joint contracture and a role for BMP signaling in tendon elongation and extracellular matrix accumulation.


Assuntos
Contratura/metabolismo , Contratura/patologia , Proteína Smad4/metabolismo , Tendões/crescimento & desenvolvimento , Animais , Desenvolvimento Ósseo , Proteínas Morfogenéticas Ósseas/metabolismo , Cartilagem/crescimento & desenvolvimento , Cartilagem/metabolismo , Linhagem da Célula , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Membro Anterior , Camundongos , Músculo Esquelético/metabolismo , Transdução de Sinais , Proteína Smad4/genética , Tendões/citologia , Tendões/embriologia , Tendões/metabolismo , Fator de Crescimento Transformador beta/metabolismo
4.
Development ; 146(20)2019 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-31540914

RESUMO

The transcription factor scleraxis (Scx) is required for tendon development; however, the function of Scx is not fully understood. Although Scx is expressed by all tendon progenitors and cells, only long tendons are disrupted in the Scx-/- mutant; short tendons appear normal and the ability of muscle to attach to skeleton is not affected. We recently demonstrated that long tendons are formed in two stages: first, by muscle anchoring to skeleton via a short tendon anlage; and second, by rapid elongation of the tendon in parallel with skeletal growth. Through lineage tracing, we extend these observations to all long tendons and show that tendon elongation is fueled by recruitment of new mesenchymal progenitors. Conditional loss of Scx in mesenchymal progenitors did not affect the first stage of anchoring; however, new cells were not recruited during elongation and long tendon formation was impaired. Interestingly, for tenocyte recruitment, Scx expression was required only in the recruited cells and not in the recruiting tendon. The phenotype of Scx mutants can thus be understood as a failure of tendon cell recruitment during tendon elongation.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Tendões/citologia , Tendões/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Movimento Celular/genética , Movimento Celular/fisiologia , Camundongos , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo
5.
Development ; 146(15)2019 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-31320326

RESUMO

Tendon and bone are attached by a transitional connective tissue that is morphologically graded from tendinous to osseous and develops from bipotent progenitors that co-express scleraxis (Scx) and Sox9 (Scx+/Sox9+). Scx+/Sox9+ progenitors have the potential to differentiate into either tenocytes or chondrocytes, yet the developmental mechanism that spatially resolves their bipotency at the tendon-bone interface during embryogenesis remains unknown. Here, we demonstrate that development of Scx+/Sox9+ progenitors within the mammalian lower jaw requires FGF signaling. We find that loss of Fgfr2 in the mouse tendon-bone interface reduces Scx expression in Scx+/Sox9+ progenitors and induces their biased differentiation into Sox9+ chondrocytes. This expansion of Sox9+ chondrocytes, which is concomitant with decreased Notch2-Dll1 signaling, prevents formation of a mixed population of chondrocytes and tenocytes, and instead results in ectopic endochondral bone at tendon-bone attachment units. Our work shows that FGF signaling directs zonal patterning at the boundary between tendon and bone by regulating cell fate decisions through a mechanism that employs Notch signaling.


Assuntos
Osso e Ossos/metabolismo , Condrócitos/citologia , Fatores de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Tendões/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Osso e Ossos/citologia , Diferenciação Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Arcada Osseodentária/metabolismo , Camundongos , Camundongos Knockout , Fatores de Transcrição SOX9/metabolismo , Transdução de Sinais/fisiologia , Células-Tronco/fisiologia , Tendões/citologia , Tenócitos/citologia
6.
Development ; 145(24)2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30504126

RESUMO

Tendon-bone attachment sites, called entheses, are essential for musculoskeletal function. They are formed embryonically by Sox9+ progenitors and continue to develop postnatally, utilizing Gli1 lineage cells. Despite their importance, we lack information on the transition from embryonic to mature enthesis and on the relation between Sox9+ progenitors and the Gli1 lineage. Here, by performing a series of lineage tracing experiments in mice, we identify the onset of Gli1 lineage contribution to different entheses. We show that Gli1 expression is regulated embryonically by SHH signaling, whereas postnatally it is maintained by IHH signaling. During bone elongation, some entheses migrate along the bone shaft, whereas others remain stationary. Interestingly, in stationary entheses Sox9+ cells differentiate into the Gli1 lineage, but in migrating entheses this lineage is replaced by Gli1 lineage. These Gli1+ progenitors are defined embryonically to occupy the different domains of the mature enthesis. Overall, these findings demonstrate a developmental strategy whereby one progenitor population establishes a simple embryonic tissue, whereas another population contributes to its maturation. Moreover, they suggest that different cell populations may be considered for cell-based therapy of enthesis injuries.


Assuntos
Osso e Ossos/fisiologia , Movimento , Células-Tronco/citologia , Tendões/fisiologia , Animais , Animais Recém-Nascidos , Compartimento Celular , Morte Celular , Linhagem da Célula , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário , Feminino , Proteínas Hedgehog/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Modelos Biológicos , Osteoclastos/citologia , Osteoclastos/metabolismo , Fagócitos/citologia , Fagócitos/metabolismo , Fatores de Transcrição SOX9/metabolismo , Células-Tronco/metabolismo , Proteína GLI1 em Dedos de Zinco/metabolismo
7.
J Biol Chem ; 293(16): 5766-5780, 2018 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-29507095

RESUMO

Tendon is a dense connective tissue that transmits high mechanical forces from skeletal muscle to bone. The transcription factor scleraxis (Scx) is a highly specific marker of both precursor and mature tendon cells (tenocytes). Mice lacking scx exhibit a specific and virtually complete loss of tendons during development. However, the functional contribution of Scx to wound healing in adult tendon has not yet been fully characterized. Here, using ScxGFP-tracking and loss-of-function systems, we show in an adult mouse model of Achilles tendon injury that paratenon cells, representing a stem cell antigen-1 (Sca-1)-positive and Scx-negative progenitor subpopulation, display Scx induction, migrate to the wound site, and produce extracellular matrix (ECM) to bridge the defect, whereas resident tenocytes exhibit a delayed response. Scx induction in the progenitors is initiated by transforming growth factor ß (TGF-ß) signaling. scx-deficient mice had migration of Sca-1-positive progenitor cell to the lesion site but impaired ECM assembly to bridge the defect. Mechanistically, scx-null progenitors displayed higher chondrogenic potential with up-regulation of SRY-box 9 (Sox9) coactivator PPAR-γ coactivator-1α (PGC-1α) in vitro, and knock-in analysis revealed that forced expression of full-length scx significantly inhibited Sox9 expression. Accordingly, scx-null wounds formed cartilage-like tissues that developed ectopic ossification. Our findings indicate a critical role of Scx in a progenitor-cell lineage in wound healing of adult mouse tendon. These progenitor cells could represent targets in strategies to facilitate tendon repair. We propose that this lineage-regulatory mechanism in tissue progenitors could apply to a broader set of tissues or biological systems in the body.


Assuntos
Tendão do Calcâneo/citologia , Tendão do Calcâneo/fisiopatologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células-Tronco/citologia , Traumatismos dos Tendões/fisiopatologia , Cicatrização , Tendão do Calcâneo/metabolismo , Tendão do Calcâneo/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Linhagem da Célula , Movimento Celular , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Deleção de Genes , Camundongos , Camundongos Transgênicos , Transdução de Sinais , Células-Tronco/metabolismo , Células-Tronco/patologia , Traumatismos dos Tendões/genética , Traumatismos dos Tendões/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Transgenes
8.
Stem Cells ; 36(4): 527-539, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29315990

RESUMO

Tendon repair is a clinical challenge because of the limited understanding on tenogenesis. The synthesis of type I collagen (Collagen I) and other extracellular matrix are essential for tendon differentiation and homeostasis. Current studies on tenogenesis focused mostly on the tenogenic transcriptional factors while the signaling controlling tenogenesis on translational level remains largely unknown. Here, we showed that mechanistic target of rapamycin (mTOR) signaling was activated by protenogenic growth factor, transforming growth factors beta1, and insulin-like growth factor-I. The expression of mTOR was upregulated during tenogenesis of mesenchymal stem cells (MSCs). Moreover, mTOR was downregulated in human tendinopathy tissues and was inactivated upon statin treatment. Both inhibition and depletion of AKT or mTOR significantly reduced type I collagen production and impaired tenogenesis of MSCs. Tendon specific-ablation of mTOR resulted in tendon defect and reduction of Collagen I. However, there is no evident downregulation of tendon associated collagens at the transcription level. Our study demonstrated that AKT-mTOR axis is a key mediator of tendon differentiation and provided a novel therapeutic target for tendinopathy and tendon injuries. Stem Cells 2018;36:527-539.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Tendões/metabolismo , Animais , Células-Tronco Mesenquimais/citologia , Camundongos , Tendões/citologia , Fator de Crescimento Transformador beta1/metabolismo
9.
Development ; 142(10): 1831-9, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25926361

RESUMO

The current view of skeletal patterning fails to explain the formation of sesamoid bones. These small bones, which facilitate musculoskeletal function, are exceptionally embedded within tendons. Although their structural design has long puzzled researchers, only a limited model for sesamoid bone development has emerged. To date, sesamoids are thought to develop inside tendons in response to mechanical signals from the attaching muscles. However, this widely accepted model has lacked substantiation. Here, we show that, contrary to the current view, in the mouse embryo the patella initially develops as a bony process at the anteriodistal surface of the femur. Later, the patella is separated from the femur by a joint formation process that is regulated by mechanical load. Concurrently, the patella becomes superficially embedded within the quadriceps tendon. At the cellular level, we show that, similar to bone eminences, the patella is formed secondarily by a distinct pool of Sox9- and Scx-positive progenitor cells. Finally, we show that TGFß signaling is necessary for the specification of patella progenitors, whereas the BMP4 pathway is required for their differentiation. These findings establish an alternative model for patella development and provide the mechanical and molecular mechanisms that underlie this process. More broadly, our finding that activation of a joint formation program can be used to switch between the formation of bony processes and of new auxiliary bones provides a new perspective on plasticity during skeletal patterning and evolution.


Assuntos
Articulações/embriologia , Articulações/metabolismo , Patela/embriologia , Patela/metabolismo , Ossos Sesamoides/embriologia , Ossos Sesamoides/metabolismo , Animais , Proteína Morfogenética Óssea 4/metabolismo , Diferenciação Celular/fisiologia , Hibridização In Situ , Articulações/citologia , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Morfogênese/genética , Morfogênese/fisiologia , Patela/citologia , Reação em Cadeia da Polimerase em Tempo Real , Ossos Sesamoides/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo
10.
Development ; 142(14): 2431-41, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-26062940

RESUMO

The long tendons of the limb extend from muscles that reside in the zeugopod (arm/leg) to their skeletal insertions in the autopod (paw). How these connections are established along the length of the limb remains unknown. Here, we show that mouse limb tendons are formed in modular units that combine to form a functional contiguous structure; in muscle-less limbs, tendons develop in the autopod but do not extend into the zeugopod, and in the absence of limb cartilage the zeugopod segments of tendons develop despite the absence of tendons in the autopod. Analyses of cell lineage and proliferation indicate that distinct mechanisms govern the growth of autopod and zeugopod tendon segments. To elucidate the integration of these autopod and zeugopod developmental programs, we re-examined early tendon development. At E12.5, muscles extend across the full length of a very short zeugopod and connect through short anlagen of tendon progenitors at the presumptive wrist to their respective autopod tendon segment, thereby initiating musculoskeletal integration. Zeugopod tendon segments are subsequently generated by proximal elongation of the wrist tendon anlagen, in parallel with skeletal growth, underscoring the dependence of zeugopod tendon development on muscles for tendon anchoring. Moreover, a subset of extensor tendons initially form as fused structures due to initial attachment of their respective wrist tendon anlage to multiple muscles. Subsequent individuation of these tendons depends on muscle activity. These results establish an integrated model for limb tendon development that provides a framework for future analyses of tendon and musculoskeletal phenotypes.


Assuntos
Extremidades/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Músculo Esquelético/embriologia , Tendões/embriologia , Animais , Apoptose , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Cartilagem/metabolismo , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Deleção de Genes , Proteínas de Fluorescência Verde/metabolismo , Articulação Metacarpofalângica/patologia , Camundongos , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Músculo Esquelético/metabolismo , Fenótipo , Fatores de Transcrição SOX9/genética , Tendões/metabolismo
11.
Development ; 141(19): 3683-96, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25249460

RESUMO

The molecular signals driving tendon development are not fully identified. We have undertaken a transcriptome analysis of mouse limb tendon cells that were isolated at different stages of development based on scleraxis (Scx) expression. Microarray comparisons allowed us to establish a list of genes regulated in tendon cells during mouse limb development. Bioinformatics analysis of the tendon transcriptome showed that the two most strongly modified signalling pathways were TGF-ß and MAPK. TGF-ß/SMAD2/3 gain- and loss-of-function experiments in mouse limb explants and mesenchymal stem cells showed that TGF-ß signalling was sufficient and required via SMAD2/3 to drive mouse mesodermal stem cells towards the tendon lineage ex vivo and in vitro. TGF-ß was also sufficient for tendon gene expression in late limb explants during tendon differentiation. FGF does not have a tenogenic effect and the inhibition of the ERK MAPK signalling pathway was sufficient to activate Scx in mouse limb mesodermal progenitors and mesenchymal stem cells.


Assuntos
Extremidades/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Transdução de Sinais/fisiologia , Tendões/citologia , Transcriptoma/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Biologia Computacional , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/genética , Hibridização In Situ , Células-Tronco Mesenquimais/metabolismo , Camundongos , Análise em Microsséries , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Tendões/metabolismo , Transcriptoma/genética , Fator de Crescimento Transformador beta/metabolismo
12.
BMC Biol ; 14: 21, 2016 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-26988708

RESUMO

BACKGROUND: Resident fibroblasts synthesize the cardiac extracellular matrix, and can undergo phenotype conversion to myofibroblasts to augment matrix production, impairing function and contributing to organ failure. A significant gap in our understanding of the transcriptional regulation of these processes exists. Given the key role of this phenotype conversion in fibrotic disease, the identification of such novel transcriptional regulators may yield new targets for therapies for fibrosis. RESULTS: Using explanted primary cardiac fibroblasts in gain- and loss-of-function studies, we found that scleraxis critically controls cardiac fibroblast/myofibroblast phenotype by direct transcriptional regulation of myriad genes that effectively define these cells, including extracellular matrix components and α-smooth muscle actin. Scleraxis furthermore potentiated the TGFß/Smad3 signaling pathway, a key regulator of myofibroblast conversion, by facilitating transcription complex formation. While scleraxis promoted fibroblast to myofibroblast conversion, loss of scleraxis attenuated myofibroblast function and gene expression. These results were confirmed in scleraxis knockout mice, which were cardiac matrix-deficient and lost ~50% of their complement of cardiac fibroblasts, with evidence of impaired epithelial-to-mesenchymal transition (EMT). Scleraxis directly transactivated several EMT marker genes, and was sufficient to induce mesenchymal/fibroblast phenotype conversion of A549 epithelial cells. Conversely, loss of scleraxis attenuated TGFß-induced EMT marker expression. CONCLUSIONS: Our results demonstrate that scleraxis is a novel and potent regulator of cellular progression along the continuum culminating in the cardiac myofibroblast phenotype. Scleraxis was both sufficient to drive conversion, and required for full conversion to occur. Scleraxis fulfills this role by direct transcriptional regulation of key target genes, and by facilitating TGFß/Smad signaling. Given the key role of fibroblast to myofibroblast conversion in fibrotic diseases in the heart and other tissue types, scleraxis may be an important target for therapeutic development.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fibroblastos/citologia , Miocárdio/citologia , Miofibroblastos/citologia , Actinas/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Fibroblastos/metabolismo , Deleção de Genes , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/metabolismo , Miofibroblastos/metabolismo , Células NIH 3T3 , Fenótipo , Ratos Sprague-Dawley , Transdução de Sinais , Proteína Smad3/metabolismo , Ativação Transcricional
13.
Development ; 137(17): 2807-17, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20699295

RESUMO

The formation of the musculoskeletal system represents an intricate process of tissue assembly involving heterotypic inductive interactions between tendons, muscles and cartilage. An essential component of all musculoskeletal systems is the anchoring of the force-generating muscles to the solid support of the organism: the skeleton in vertebrates and the exoskeleton in invertebrates. Here, we discuss recent findings that illuminate musculoskeletal assembly in the vertebrate embryo, findings that emphasize the reciprocal interactions between the forming tendons, muscle and cartilage tissues. We also compare these events with those of the corresponding system in the Drosophila embryo, highlighting distinct and common pathways that promote efficient locomotion while preserving the form of the organism.


Assuntos
Desenvolvimento Musculoesquelético , Tendões/embriologia , Animais , Drosophila/embriologia , Drosophila/genética , Drosophila/fisiologia , Músculos/embriologia , Músculos/fisiologia , Desenvolvimento Musculoesquelético/genética , Desenvolvimento Musculoesquelético/fisiologia , Transdução de Sinais , Tendões/fisiologia , Vertebrados/embriologia , Vertebrados/genética , Vertebrados/fisiologia
14.
Development ; 137(7): 1181-8, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20215352

RESUMO

Integration of muscle, connective tissue and skeletal patterning during development is essential for proper functioning of the musculoskeletal system. How this integration is achieved is poorly understood. There is ample evidence suggesting that skeletal pattern is programmed autonomously, whereas muscle pattern is, for the most part, programmed non-cell-autonomously. Connective tissues depend upon both muscle and skeletal tissues for their proper survival and development. Here, we employed a novel approach to dissect the coordination of musculoskeletal patterning during mouse limb development. Using both conditional gain- and loss-of-function approaches, we selectively deleted or activated the LIM-homeodomain transcription factor Lmx1b in skeletal progenitors using a Sox9-Cre knock-in allele. As Lmx1b is both necessary and sufficient to specify dorsal pattern, this approach allowed us to investigate the effect of selectively deleting or activating Lmx1b in skeletal progenitors on muscle, connective and skeletal tissues during limb development. Our results indicate that whereas Lmx1b activity is required autonomously in skeletal progenitors to direct dorsal pattern, loss or gain of Lmx1b activity in skeletal progenitors has no effect on muscle or connective tissue patterning. Hence, we show for the first time that skeletal and connective tissue patterning can be uncoupled, indicating a degree of autonomy in the formation of the musculoskeletal system.


Assuntos
Padronização Corporal/fisiologia , Cartilagem/citologia , Extremidades , Proteínas de Homeodomínio/metabolismo , Células-Tronco/fisiologia , Fatores de Transcrição/metabolismo , Animais , Animais Recém-Nascidos , Osso e Ossos/citologia , Osso e Ossos/fisiologia , Linhagem da Célula , Tecido Conjuntivo/anatomia & histologia , Tecido Conjuntivo/fisiologia , Extremidades/anatomia & histologia , Extremidades/embriologia , Técnicas de Introdução de Genes , Proteínas de Homeodomínio/genética , Articulações/anatomia & histologia , Articulações/embriologia , Articulações/metabolismo , Proteínas com Homeodomínio LIM , Mesoderma/citologia , Camundongos , Camundongos Transgênicos , Músculo Esquelético/citologia , Músculo Esquelético/fisiologia , Fenótipo , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Células-Tronco/citologia , Fatores de Transcrição/genética
15.
Nat Methods ; 5(7): 609-12, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18516047

RESUMO

Quantitative mapping of the normal tissue dynamics of an entire developing mammalian organ has not been achieved so far but is essential to understand developmental processes and to provide quantitative data for computational modeling. We developed a four-dimensional (4D) imaging technique that can be used to quantitatively image tissue movements and dynamic GFP expression domains in a growing transgenic mouse limb by time-lapse optical projection tomography (OPT).


Assuntos
Extremidades/embriologia , Imageamento Tridimensional/métodos , Tomografia Óptica/métodos , Animais , Padronização Corporal , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Técnicas In Vitro , Camundongos , Camundongos Transgênicos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fatores de Tempo
16.
Dev Dyn ; 239(11): 2898-910, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20865781

RESUMO

The myotome is a segmented paraxial muscle present in all early vertebrate embryos, which in amniotes disappears in mid-embryogenesis, and is replaced by complex epaxial and hypaxial musculature. Little is known about how this transition occurs. Here, we describe the detailed morphogenesis of the epaxial muscles from the epaxial myotome, in rodent embryos. The results show there is no apoptosis of myotomal fibres during the transition, and that the epaxial muscles arise by translocation, re-orientation, and elongation of the myotomal myocytes followed by cleavage of the myotomal masses. Myotomal myocytes transit from a mononucleated to a multinucleated state just before onset of this transformation. Each newly-formed epaxial muscle anlagen includes populations of Pax3- and Pax7-positive muscle progenitors, with different distributions. Using transgenic mouse embryos bearing a GFP marker for Scleraxis, we show that tendon progenitors are tightly associated with the sides and ends of myotomal myocytes as they re-orient and elongate.


Assuntos
Embrião de Mamíferos/metabolismo , Morfogênese/fisiologia , Desenvolvimento Muscular/fisiologia , Animais , Apoptose/genética , Apoptose/fisiologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Embrião de Mamíferos/citologia , Feminino , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Morfogênese/genética , Células Musculares/citologia , Células Musculares/metabolismo , Desenvolvimento Muscular/genética , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/metabolismo , Gravidez , Ratos , Ratos Wistar
17.
Stem Cells Dev ; 30(11): 601-609, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33757300

RESUMO

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


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste , Osteogênese , Diferenciação Celular/genética , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Mesoderma , Osteogênese/genética , Tendões
18.
Stem Cell Reports ; 16(12): 2942-2957, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34822771

RESUMO

Understanding cell recruitment in damaged tendons is critical for improvements in regenerative therapy. We recently reported that targeted disruption of transforming growth factor beta (TGFß) type II receptor in the tendon cell lineage (Tgfbr2ScxCre) resulted in resident tenocyte dedifferentiation and tendon deterioration in postnatal stages. Here we extend the analysis and identify direct recruitment of stem/progenitor cells into the degenerative mutant tendons. Cre-mediated lineage tracing indicates that these cells are not derived from tendon-ensheathing tissues or from a Scleraxis-expressing lineage, and they turned on tendon markers only upon entering the mutant tendons. Through immunohistochemistry and inducible gene deletion, we further find that the recruited cells originated from a Sox9-expressing lineage and their recruitment was dependent on cell autonomous TGFß signaling. The cells identified in this study thus differ from previous reports of cell recruitment into injured tendons and suggest a critical role for TGFß signaling in cell recruitment, providing insights that may support improvements in tendon repair.


Assuntos
Transdução de Sinais , Células-Tronco/metabolismo , Tendões/patologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Biomarcadores/metabolismo , Células Cultivadas , Células Clonais , Proteínas de Fluorescência Verde/metabolismo , Integrases/metabolismo , Camundongos , Modelos Biológicos , Mutação/genética , Receptor do Fator de Crescimento Transformador beta Tipo II/metabolismo , Tendões/ultraestrutura , Fatores de Tempo
19.
Nat Commun ; 12(1): 3851, 2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34158501

RESUMO

Positional information driving limb muscle patterning is contained in connective tissue fibroblasts but not in myogenic cells. Limb muscles originate from somites, while connective tissues originate from lateral plate mesoderm. With cell and genetic lineage tracing we challenge this model and identify an unexpected contribution of lateral plate-derived fibroblasts to the myogenic lineage, preferentially at the myotendinous junction. Analysis of single-cell RNA-sequencing data from whole limbs at successive developmental stages identifies a population displaying a dual muscle and connective tissue signature. BMP signalling is active in this dual population and at the tendon/muscle interface. In vivo and in vitro gain- and loss-of-function experiments show that BMP signalling regulates a fibroblast-to-myoblast conversion. These results suggest a scenario in which BMP signalling converts a subset of lateral plate mesoderm-derived cells to a myogenic fate in order to create a boundary of fibroblast-derived myonuclei at the myotendinous junction that controls limb muscle patterning.


Assuntos
Padronização Corporal/genética , Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Músculo Esquelético/metabolismo , Somitos/metabolismo , Animais , Linhagem da Célula/genética , Células Cultivadas , Embrião de Galinha , Extremidades/embriologia , Fibroblastos/citologia , Mesoderma/citologia , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Desenvolvimento Muscular/genética , Músculo Esquelético/citologia , Músculo Esquelético/embriologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Somitos/citologia , Somitos/embriologia
20.
Elife ; 92020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31961320

RESUMO

Studies of cell fate focus on specification, but little is known about maintenance of the differentiated state. In this study, we find that the mouse tendon cell fate requires continuous maintenance in vivo and identify an essential role for TGFß signaling in maintenance of the tendon cell fate. To examine the role of TGFß signaling in tenocyte function the TGFß type II receptor (Tgfbr2) was targeted in the Scleraxis-expressing cell lineage using the ScxCre deletor. Tendon development was not disrupted in mutant embryos, but shortly after birth tenocytes lost differentiation markers and reverted to a more stem/progenitor state. Viral reintroduction of Tgfbr2 to mutants prevented and even rescued tenocyte dedifferentiation suggesting a continuous and cell autonomous role for TGFß signaling in cell fate maintenance. These results uncover the critical importance of molecular pathways that maintain the differentiated cell fate and a key role for TGFß signaling in these processes.


Assuntos
Receptor do Fator de Crescimento Transformador beta Tipo II/metabolismo , Tendões/citologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Desdiferenciação Celular , Linhagem da Célula , Regulação da Expressão Gênica , Camundongos , Mutação , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/metabolismo , Tendões/metabolismo , Tenócitos/citologia , Tenócitos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA