Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.891
Filtrar
1.
Dev Biol ; 482: 91-100, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34929174

RESUMO

Ciliogenic components, such as the family of intraflagellar transport (IFT) proteins, are recognized to play key roles in endochondral ossification, a critical process to form most bones. However, the unique functions and roles of each IFT during endochondral ossification remain unclear. Here, we show that IFT20 is required for endochondral ossification in mice. Utilizing osteo-chondrocyte lineage-specific Cre mice (Prx1-Cre and Col2-Cre), we deleted Ift20 to examine its function. Although chondrocyte-specific Ift20 deletion with Col2-Cre mice did not cause any overt skeletal defects, mesoderm-specific Ift20 deletion using Prx1-Cre (Ift20:Prx1-Cre) mice resulted in shortened limb outgrowth. Primary cilia were absent on chondrocytes of Ift20:Prx1-Cre mice, and ciliary-mediated Hedgehog signaling was attenuated in Ift20:Prx1-Cre mice. Interestingly, loss of Ift20 also increased Fgf18 expression in the perichondrium that sustained Sox9 expression, thus preventing endochondral ossification. Inhibition of enhanced phospho-ERK1/2 activation partially rescued defective chondrogenesis in Ift20 mutant cells, supporting an important role for FGF signaling. Our findings demonstrate that IFT20 is a critical regulator of temporospatial FGF signaling that is required for endochondral ossification.


Assuntos
Proteínas de Transporte/genética , Condrogênese/genética , Cílios/genética , Deformidades Congênitas dos Membros/genética , Osteogênese/genética , Animais , Osso e Ossos/embriologia , Cartilagem/embriologia , Diferenciação Celular/genética , Condrócitos/fisiologia , Condrogênese/fisiologia , Proteínas Cromossômicas não Histona/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Camundongos , Camundongos Knockout , Osteogênese/fisiologia , Fatores de Transcrição SOX9/biossíntese , Transdução de Sinais/fisiologia
2.
Commun Biol ; 4(1): 658, 2021 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-34079065

RESUMO

It has not been well studied which cells and related mechanisms contribute to endochondral ossification. Here, we fate mapped the leptin receptor-expressing (LepR+) mesenchymal stem cells (MSCs) in different embryonic and adult extremities using Lepr-cre; tdTomato mice and investigated the underling mechanism using Lepr-cre; Ppp2r1afl/fl mice. Tomato+ cells appear in the primary and secondary ossification centers and express the hypertrophic markers. Ppp2r1a deletion in LepR+ MSCs reduces the expression of Runx2, Osterix, alkaline phosphatase, collagen X, and MMP13, but increases that of the mature adipocyte marker perilipin, thereby reducing trabecular bone density and enhancing fat content. Mechanistically, PP2A dephosphorylates Runx2 and BRD4, thereby playing a major role in positively and negatively regulating osteogenesis and adipogenesis, respectively. Our data identify LepR+ MSC as the cell origin of endochondral ossification during embryonic and postnatal bone growth and suggest that PP2A is a therapeutic target in the treatment of dysregulated bone formation.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteogênese/fisiologia , Proteína Fosfatase 2/metabolismo , Receptores para Leptina/metabolismo , Adipogenia , Animais , Densidade Óssea , Osso e Ossos/citologia , Osso e Ossos/embriologia , Osso e Ossos/metabolismo , Diferenciação Celular , Proliferação de Células , Condrogênese , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas Nucleares/metabolismo , Fosforilação , Gravidez , Proteína Fosfatase 2/deficiência , Proteína Fosfatase 2/genética , Fatores de Transcrição/metabolismo
3.
Sci Rep ; 11(1): 10933, 2021 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-34035368

RESUMO

The human fetal osteoblast cell line (hFOB 1.19) has been proposed as an accessible experimental model for study of osteoblast biology relating to drug development and biomaterial engineering. For their multilineage differentiation potential, hFOB has been compared to human mesenchymal progenitor cells and used to investigate bone-metabolism in vitro. Hereby, we studied whether and to what extent the conditionally immortalized cell line hFOB 1.19 can serve as a surrogate model for bone-marrow derived mesenchymal stromal cells (bmMSC). hFOB indeed exhibit specific characteristics reminiscent of bmMSC, as colony formation, migration capacity and the propensity to grow as multicellular aggregates. After prolonged culture, in contrast to the expected effect of immortalization, hFOB acquired a delayed growth rate. In close resemblance to bmMSC at increasing passages, also hFOB showed morphological abnormalities, enlargement and finally reduced proliferation rates together with enhanced expression of the cell cycle inhibitors p21 and p16. hFOB not only have the ability to undergo multilineage differentiation but portray several important aspects of human bone marrow mesenchymal stromal cells. Superior to primary MSC and osteoblasts, hFOB enabled the generation of continuous cell lines. These provide an advanced basis for investigating age-related dysfunctions of MSCs in an in vitro 3D-stem cell microenvironment.


Assuntos
Osso e Ossos/embriologia , Técnicas de Cultura de Células/métodos , Células-Tronco Mesenquimais/citologia , Osteoblastos/citologia , Osso e Ossos/citologia , Osso e Ossos/metabolismo , Diferenciação Celular , Linhagem Celular , Movimento Celular , Proliferação de Células , Meios de Cultivo Condicionados , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/metabolismo
4.
Development ; 148(10)2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-34042967

RESUMO

Regeneration as an adult developmental process is in many aspects similar to embryonic development. Although many studies point out similarities and differences, no large-scale, direct and functional comparative analyses between development and regeneration of a specific cell type or structure in one animal exist. Here, we use the brittle star Amphiura filiformis to characterise the role of the FGF signalling pathway during skeletal development in embryos and arm regeneration. In both processes, we find ligands expressed in ectodermal cells that flank underlying skeletal mesenchymal cells, which express the receptors. Perturbation of FGF signalling showed inhibited skeleton formation in both embryogenesis and regeneration, without affecting other key developmental processes. Differential transcriptome analysis finds mostly differentiation genes rather than transcription factors to be downregulated in both contexts. Moreover, comparative gene analysis allowed us to discover brittle star-specific differentiation genes. In conclusion, our results show that the FGF pathway is crucial for skeletogenesis in the brittle star, as in other deuterostomes, and provide evidence for the re-deployment of a developmental gene regulatory module during regeneration.


Assuntos
Desenvolvimento Ósseo/fisiologia , Regeneração Óssea/fisiologia , Osso e Ossos/embriologia , Fatores de Crescimento de Fibroblastos/metabolismo , Estrelas-do-Mar/embriologia , Animais , Osso e Ossos/metabolismo , Ectoderma/citologia , Ectoderma/metabolismo , Desenvolvimento Embrionário/genética , Extremidades/crescimento & desenvolvimento , Mesoderma/citologia , Mesoderma/metabolismo , Pirróis/farmacologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Transdução de Sinais/fisiologia , Estrelas-do-Mar/genética , Estrelas-do-Mar/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
5.
Int J Mol Sci ; 22(8)2021 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-33924333

RESUMO

Bone formation starts near the end of the embryonic stage of development and continues throughout life during bone modeling and growth, remodeling, and when needed, regeneration. Bone-forming cells, traditionally termed osteoblasts, produce, assemble, and control the mineralization of the type I collagen-enriched bone matrix while participating in the regulation of other cell processes, such as osteoclastogenesis, and metabolic activities, such as phosphate homeostasis. Osteoblasts are generated by different cohorts of skeletal stem cells that arise from different embryonic specifications, which operate in the pre-natal and/or adult skeleton under the control of multiple regulators. In this review, we briefly define the cellular identity and function of osteoblasts and discuss the main populations of osteoprogenitor cells identified to date. We also provide examples of long-known and recently recognized regulatory pathways and mechanisms involved in the specification of the osteogenic lineage, as assessed by studies on mice models and human genetic skeletal diseases.


Assuntos
Osteoblastos/citologia , Osteogênese , Células-Tronco/citologia , Animais , Osso e Ossos/citologia , Osso e Ossos/embriologia , Epigênese Genética , Humanos , Osteogênese/genética , Transdução de Sinais
6.
PLoS One ; 16(2): e0240642, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33626093

RESUMO

The presented experiment focuses on assessing the impact of HMB (hydroxy-ß-methobutyrate) supplementation of mothers during pregnancy on the development of the skeletal system of their offspring. For this purpose, an experiment was carried out on 12 clinically healthy sows of the Great White Poland breed, which were divided randomly into two groups the control and the HMB group. All animals were kept under standard conditions and received the same feed for pregnant females. In contrast, females from the HMB group between 70 and 90 days were supplemented with 3-hydroxy-3-methylbutyle in the amount of 0.2g/kg b.w/day. Immediately after birth, the piglets were also divided into groups based on: sex, and presence or lack HMB supplementation, and subsequently were euthanized and humerus bones from all piglets were collected. Mother's HMB supplementation during pregnancy affected the multiple index of their offspring. The higher humerus mass and length was observed with the greater effect in males. Maternal supplementation also influenced on the geometrical and mechanical properties of the humerus as in the case of mass, this effect was higher in males. Also, the collagen structure of the compacted and trabecular bone changed under the HMB addition. Maternal supplementation also affected the expression of selected proteins in growth cartilage and trabecular bone. The obtained results show that the administration to the mother during pregnancy by the HMB significantly affects the development of the humerus in many ways. The obtained results also confirm the utility of such experiments in understanding of the importance of the pregnancy diet as an develop and adaptable factor of offspring organisms and are the base for further research in that area as well as in the protein markers expression area.


Assuntos
Úmero/efeitos dos fármacos , Suínos/embriologia , Valeratos/farmacologia , Ração Animal/análise , Fenômenos Fisiológicos da Nutrição Animal , Animais , Animais Recém-Nascidos/embriologia , Animais Recém-Nascidos/metabolismo , Proteína Morfogenética Óssea 2/metabolismo , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/embriologia , Cartilagem , Dieta/veterinária , Suplementos Nutricionais , Feminino , Úmero/embriologia , Masculino , Exposição Materna , Metaloproteinase 13 da Matriz/metabolismo , Polônia , Gravidez , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Valeratos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Cell ; 184(4): 899-911.e13, 2021 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-33545089

RESUMO

Changes in appendage structure underlie key transitions in vertebrate evolution. Addition of skeletal elements along the proximal-distal axis facilitated critical transformations, including the fin-to-limb transition that permitted generation of diverse modes of locomotion. Here, we identify zebrafish mutants that form supernumerary long bones in their pectoral fins. These new bones integrate into musculature, form joints, and articulate with neighboring elements. This phenotype is caused by activating mutations in previously unrecognized regulators of appendage patterning, vav2 and waslb, that function in a common pathway. This pathway is required for appendage development across vertebrates, and loss of Wasl in mice causes defects similar to those seen in murine Hox mutants. Concordantly, formation of supernumerary bones requires Hox11 function, and mutations in the vav2/wasl pathway drive enhanced expression of hoxa11b, indicating developmental homology with the forearm. Our findings reveal a latent, limb-like pattern ability in fins that is activated by simple genetic perturbation.


Assuntos
Osso e Ossos/embriologia , Extremidades/embriologia , Peixe-Zebra/embriologia , Actinas/metabolismo , Nadadeiras de Animais/embriologia , Animais , Sequência de Bases , Padronização Corporal , Sistemas CRISPR-Cas/genética , Linhagem da Célula , Epistasia Genética , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Genes Reporter , Células HeLa , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Mutação/genética , Fenótipo , Filogenia , Transdução de Sinais/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
8.
Int J Mol Sci ; 22(3)2021 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-33573345

RESUMO

Skeletal disorders, such as osteoarthritis and bone fractures, are among the major conditions that can compromise the quality of daily life of elderly individuals. To treat them, regenerative therapies using skeletal cells have been an attractive choice for patients with unmet clinical needs. Currently, there are two major strategies to prepare the cell sources. The first is to use induced pluripotent stem cells (iPSCs) or embryonic stem cells (ESCs), which can recapitulate the skeletal developmental process and differentiate into various skeletal cells. Skeletal tissues are derived from three distinct origins: the neural crest, paraxial mesoderm, and lateral plate mesoderm. Thus, various protocols have been proposed to recapitulate the sequential process of skeletal development. The second strategy is to extract stem cells from skeletal tissues. In addition to mesenchymal stem/stromal cells (MSCs), multiple cell types have been identified as alternative cell sources. These cells have distinct multipotent properties allowing them to differentiate into skeletal cells and various potential applications for skeletal regeneration. In this review, we summarize state-of-the-art research in stem cell differentiation based on the understanding of embryogenic skeletal development and stem cells existing in skeletal tissues. We then discuss the potential applications of these cell types for regenerative medicine.


Assuntos
Desenvolvimento Ósseo/fisiologia , Osso e Ossos/fisiologia , Fraturas Ósseas/terapia , Osteoartrite/terapia , Medicina Regenerativa/métodos , Animais , Osso e Ossos/embriologia , Osso e Ossos/lesões , Diferenciação Celular/fisiologia , Modelos Animais de Doenças , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário/fisiologia , Células-Tronco Embrionárias/fisiologia , Fraturas Ósseas/fisiopatologia , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Células-Tronco Mesenquimais/fisiologia , Mesoderma/embriologia , Crista Neural/embriologia , Osteoartrite/fisiopatologia , Osteoblastos/fisiologia , Osteoblastos/transplante , Medicina Regenerativa/tendências , Transplante de Células-Tronco/métodos , Transplante de Células-Tronco/tendências
9.
Development ; 148(2)2021 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-33462117

RESUMO

The regulated expansion of chondrocytes within growth plates and joints ensures proper skeletal development through adulthood. Mutations in the transcription factor NKX3.2 underlie spondylo-megaepiphyseal-metaphyseal dysplasia (SMMD), which is characterized by skeletal defects including scoliosis, large epiphyses, wide growth plates and supernumerary distal limb joints. Whereas nkx3.2 knockdown zebrafish and mouse Nkx3.2 mutants display embryonic lethal jaw joint fusions and skeletal reductions, respectively, they lack the skeletal overgrowth seen in SMMD patients. Here, we report adult viable nkx3.2 mutant zebrafish displaying cartilage overgrowth in place of a missing jaw joint, as well as severe dysmorphologies of the facial skeleton, skullcap and spine. In contrast, cartilage overgrowth and scoliosis are absent in rare viable nkx3.2 knockdown animals that lack jaw joints, supporting post-embryonic roles for Nkx3.2. Single-cell RNA-sequencing and in vivo validation reveal increased proliferation and upregulation of stress-induced pathways, including prostaglandin synthases, in mutant chondrocytes. By generating a zebrafish model for the skeletal overgrowth defects of SMMD, we reveal post-embryonic roles for Nkx3.2 in dampening proliferation and buffering the stress response in joint-associated chondrocytes.


Assuntos
Osso e Ossos/embriologia , Osso e Ossos/metabolismo , Proteínas de Homeodomínio/metabolismo , Osteocondrodisplasias/embriologia , Fatores de Transcrição/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Cartilagem/embriologia , Cartilagem/patologia , Condrócitos/metabolismo , Modelos Animais de Doenças , Embrião não Mamífero/anormalidades , Embrião não Mamífero/patologia , Regulação da Expressão Gênica no Desenvolvimento , Arcada Osseodentária/embriologia , Arcada Osseodentária/patologia , Articulações/anormalidades , Articulações/embriologia , Articulações/patologia , Mitose/genética , Morfolinos/farmacologia , Mutação/genética , RNA-Seq , Análise de Célula Única , Crânio/anormalidades , Crânio/embriologia , Crânio/patologia , Coluna Vertebral/anormalidades , Coluna Vertebral/embriologia , Coluna Vertebral/patologia , Estresse Fisiológico/genética , Regulação para Cima/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
10.
J Dev Orig Health Dis ; 12(3): 384-395, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32500846

RESUMO

Adverse programming of adult non-communicable disease can be induced by poor maternal nutrition during pregnancy and the periconception period has been identified as a vulnerable period. In the current study, we used a mouse maternal low-protein diet fed either for the duration of pregnancy (LPD) or exclusively during the preimplantation period (Emb-LPD) with control nutrition provided thereafter and postnatally to investigate effects on fetal bone development and quality. This model has been shown previously to induce cardiometabolic and neurological disease phenotypes in offspring. Micro 3D computed tomography examination at fetal stages Embryonic day E14.5 and E17.4, reflecting early and late stages of bone formation, demonstrated LPD treatment caused increased bone formation of relative high mineral density quality in males, but not females, at E14.5, disproportionate to fetal growth, with bone quality maintained at E17.5. In contrast, Emb-LPD caused a late increase in male fetal bone growth, proportionate to fetal growth, at E17.5, affecting central and peripheral skeleton and of reduced mineral density quality relative to controls. These altered dynamics in bone growth coincide with increased placental efficiency indicating compensatory responses to dietary treatments. Overall, our data show fetal bone formation and mineral quality is dependent upon maternal nutritional protein content and is sex-specific. In particular, we find the duration and timing of poor maternal diet to be critical in the outcomes with periconceptional protein restriction leading to male offspring with increased bone growth but of poor mineral density, thereby susceptible to later disease risk.


Assuntos
Densidade Óssea , Osso e Ossos/embriologia , Dieta com Restrição de Proteínas/efeitos adversos , Desenvolvimento Fetal , Fenômenos Fisiológicos da Nutrição Materna , Animais , Feminino , Masculino , Camundongos , Gravidez
11.
Development ; 148(1)2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33298461

RESUMO

Vertebrate axial skeletal patterning is controlled by co-linear expression of Hox genes and axial level-dependent activity of HOX protein combinations. MEIS transcription factors act as co-factors of HOX proteins and profusely bind to Hox complex DNA; however, their roles in mammalian axial patterning remain unknown. Retinoic acid (RA) is known to regulate axial skeletal element identity through the transcriptional activity of its receptors; however, whether this role is related to MEIS/HOX activity remains unknown. Here, we study the role of Meis in axial skeleton formation and its relationship to the RA pathway in mice. Meis elimination in the paraxial mesoderm produces anterior homeotic transformations and rib mis-patterning associated to alterations of the hypaxial myotome. Although Raldh2 and Meis positively regulate each other, Raldh2 elimination largely recapitulates the defects associated with Meis deficiency, and Meis overexpression rescues the axial skeletal defects in Raldh2 mutants. We propose a Meis-RA-positive feedback loop, the output of which is Meis levels, that is essential to establish anterior-posterior identities and patterning of the vertebrate axial skeleton.


Assuntos
Padronização Corporal , Osso e Ossos/embriologia , Osso e Ossos/metabolismo , Retroalimentação Fisiológica , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Proteína Meis1/metabolismo , Tretinoína/metabolismo , Aldeído Oxirredutases/deficiência , Aldeído Oxirredutases/metabolismo , Alelos , Animais , Padronização Corporal/genética , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário/genética , Feto/metabolismo , Proteínas de Homeodomínio/genética , Integrases/metabolismo , Camundongos , Modelos Biológicos , Mutação/genética , Proteína Meis1/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais
12.
Dev Dyn ; 250(3): 414-449, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33314394

RESUMO

Skeletal elements have a diverse range of shapes and sizes specialized to their various roles including protecting internal organs, locomotion, feeding, hearing, and vocalization. The precise positioning, size, and shape of skeletal elements is therefore critical for their function. During embryonic development, bone forms by endochondral or intramembranous ossification and can arise from the paraxial and lateral plate mesoderm or neural crest. This review describes inductive mechanisms to position and pattern bones within the developing embryo, compares and contrasts the intrinsic vs extrinsic mechanisms of endochondral and intramembranous skeletal development, and details known cellular processes that precisely determine skeletal shape and size. Key cellular mechanisms are employed at distinct stages of ossification, many of which occur in response to mechanical cues (eg, joint formation) or preempting future load-bearing requirements. Rapid shape changes occur during cellular condensation and template establishment. Specialized cellular behaviors, such as chondrocyte hypertrophy in endochondral bone and secondary cartilage on intramembranous bones, also dramatically change template shape. Once ossification is complete, bone shape undergoes functional adaptation through (re)modeling. We also highlight how alterations in these cellular processes contribute to evolutionary change and how differences in the embryonic origin of bones can influence postnatal bone repair.


Assuntos
Osso e Ossos/embriologia , Diferenciação Celular , Condrócitos/metabolismo , Condrogênese , Osteoblastos/metabolismo , Osteogênese/fisiologia , Animais , Humanos
13.
Pediatr Radiol ; 50(12): 1650-1657, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33135135

RESUMO

Skeletal dysplasias have been recognised since recorded history began. The advent of radiography at the beginning of the 20th century and the subsequent introduction of departments of radiology have had tremendous impact and allowed conditions to be identified by their specific radiographic phenotypes. This has been enhanced by the addition of cross-sectional modalities (ultrasound, computed tomography and magnetic resonance imaging), which have allowed for prenatal recognition and diagnosis of skeletal dysplasias, and by the recent explosion in identified genes. There are more than 400 recognised skeletal dysplasias, many of which (due to their rarity) the practising clinician (radiologist, paediatrician, geneticist) may never come across. This article provides a historical overview of aids to the radiologic diagnosis of skeletal dysplasias.


Assuntos
Doenças do Desenvolvimento Ósseo/diagnóstico por imagem , Diagnóstico por Imagem/métodos , Diagnóstico Pré-Natal/métodos , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/embriologia , Diagnóstico por Imagem/tendências , Feminino , Humanos , Gravidez , Diagnóstico Pré-Natal/tendências
14.
Development ; 147(21)2020 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-32994166

RESUMO

The Hippo-YAP/TAZ pathway is an important regulator of tissue growth, but can also control cell fate or tissue morphogenesis. Here, we investigate the function of the Hippo pathway during the development of cartilage, which forms the majority of the skeleton. Previously, YAP was proposed to inhibit skeletal size by repressing chondrocyte proliferation and differentiation. We find that, in vitro, Yap/Taz double knockout impairs murine chondrocyte proliferation, whereas constitutively nuclear nls-YAP5SA accelerates proliferation, in line with the canonical role of this pathway in most tissues. However, in vivo, cartilage-specific knockout of Yap/Taz does not prevent chondrocyte proliferation, differentiation or skeletal growth, but rather results in various skeletal deformities including cleft palate. Cartilage-specific expression of nls-YAP5SA or knockout of Lats1/2 do not increase cartilage growth, but instead lead to catastrophic malformations resembling chondrodysplasia or achondrogenesis. Physiological YAP target genes in cartilage include Ctgf, Cyr61 and several matrix remodelling enzymes. Thus, YAP/TAZ activity controls chondrocyte proliferation in vitro, possibly reflecting a regenerative response, but is dispensable for chondrocyte proliferation in vivo, and instead functions to control cartilage morphogenesis via regulation of the extracellular matrix.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Osso e Ossos/embriologia , Osso e Ossos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Morfogênese , Proteínas Serina-Treonina Quinases/metabolismo , Transativadores/metabolismo , Animais , Osso e Ossos/anormalidades , Osso e Ossos/patologia , Cartilagem/patologia , Núcleo Celular/metabolismo , Proliferação de Células , Condrócitos/metabolismo , Condrócitos/patologia , Fissura Palatina/patologia , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Lâmina de Crescimento/patologia , Via de Sinalização Hippo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfogênese/genética , Transdução de Sinais , Proteínas Supressoras de Tumor/metabolismo , Proteínas de Sinalização YAP
15.
Reprod Domest Anim ; 55(9): 1180-1189, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32608107

RESUMO

Primiparous ewes and multiparous ewes show physiological differences during pregnancy, which can have an impact on the development of their offspring. The objective of this study was to compare the changes in the metabolic profile and in the size of some foetal bones throughout gestation between primiparous and multiparous ewes. Twelve primiparous (PM) ewes and 14 multiparous (MT) ewes were used. According to the dates of lambing, two groups of ewes were formed: Group 1 (G1, n = 6 PM and n = 7 MT) and Group 2 (G2, n = 6 PM and n = 7 MT). The body weight, body condition score, metabolic and foetal morphometric parameters were determined from before conception until the end of gestation. After lambing, the body weight and survival rate during the first 72 hr of life of lambs, as well as the maternal behaviour score were recorded. The PM ewes were lighter (p < .01) and had a greater mobilization of body reserves during gestation, mainly evidenced by a greater serum concentration of NEFAs and lower serum concentration of total proteins (p < .05) compared with the MT ewes. The parity did not affect the foetal morphometric variables. The lambs of MT ewes were heavier at parturition (p = .002) and tended to have a greater survival rate than those lambs of PM ewes (p = .09). In conclusion, PM ewes and MT ewes differ in their metabolic profile throughout the gestation. However, in the present study, we did not find parity differences in the dimensions of foetal bones during growth in gestation.


Assuntos
Osso e Ossos/embriologia , Paridade , Gravidez/metabolismo , Carneiro Doméstico/embriologia , Carneiro Doméstico/metabolismo , Animais , Animais Recém-Nascidos , Peso Corporal , Desenvolvimento Ósseo , Feminino , Desenvolvimento Fetal , Metaboloma , Gravidez/fisiologia
16.
Biol Rev Camb Philos Soc ; 95(4): 986-1019, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32338826

RESUMO

This review describes the formation, structure, and function of bony compartments in antlers, horns, ossicones, osteoderm and the os penis/os clitoris (collectively referred to herein as AHOOO structures) in extant mammals. AHOOOs are extra-skeletal bones that originate from subcutaneous (dermal) tissues in a wide variety of mammals, and this review elaborates on the co-development of the bone and skin in these structures. During foetal stages, primordial cells for the bony compartments arise in subcutaneous tissues. The epithelial-mesenchymal transition is assumed to play a key role in the differentiation of bone, cartilage, skin and other tissues in AHOOO structures. AHOOO ossification takes place after skeletal bone formation, and may depend on sexual maturity. Skin keratinization occurs in tandem with ossification and may be under the control of androgens. Both endochondral and intramembranous ossification participate in bony compartment formation. There is variation in gradients of density in different AHOOO structures. These gradients, which vary according to function and species, primarily reduce mechanical stress. Anchorage of AHOOOs to their surrounding tissues fortifies these structures and is accomplished by bone-bone fusion and Sharpey fibres. The presence of the integument is essential for the protection and function of the bony compartments. Three major functions can be attributed to AHOOOs: mechanical, visual, and thermoregulatory. This review provides the first extensive comparative description of the skeletal and integumentary systems of AHOOOs in a variety of mammals.


Assuntos
Chifres de Veado/fisiologia , Cervos/anatomia & histologia , Cornos/fisiologia , Mamíferos/anatomia & histologia , Mamíferos/fisiologia , Ruminantes/anatomia & histologia , Animais , Chifres de Veado/anatomia & histologia , Chifres de Veado/crescimento & desenvolvimento , Tatus/anatomia & histologia , Tatus/crescimento & desenvolvimento , Osso e Ossos/embriologia , Clitóris/anatomia & histologia , Cervos/crescimento & desenvolvimento , Feminino , Girafas/anatomia & histologia , Girafas/crescimento & desenvolvimento , Cornos/anatomia & histologia , Cornos/crescimento & desenvolvimento , Masculino , Mamíferos/crescimento & desenvolvimento , Pênis/anatomia & histologia , Ruminantes/crescimento & desenvolvimento , Pele/embriologia , Pele/crescimento & desenvolvimento , Tela Subcutânea/anatomia & histologia , Tela Subcutânea/embriologia
17.
J Anat ; 237(1): 105-118, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32255516

RESUMO

Studying the timing of the main events of embryonic and fetal development may clarify the strategies adopted by species to maximize neonatal survival and the consequences of these events for their life history. This study describes bone development during the fetal phase of the lowland paca (Cuniculus paca), comparing it with other precocial or altricial species, and its relationship with the species' adaptive strategies. A total of 102 embryos/fetuses obtained over the course of 17 years through collaboration with local subsistence hunters in the Amazon were analyzed. Measurements of mineralization of the axial and appendicular skeletons were performed by ultrasonography using a 10-18-MHz linear transducer. The chronological order of occurrence of mineralization in relation to the total dorsal length (TDL) was: skull (TDL = 4.1 cm); vertebral bodies (TDL = 4.6 cm); scapula, humerus, radius, ulna, ilium, ischium, femur, tibia, and fibula (TDL = 6.7 cm); ribs (TDL = 7.8 cm); clavicle (TDL = 8.5 cm); metacarpi/metatarsi (TDL = 11 cm); phalanges (TDL = 15 cm); tarsus (TDL = 18 cm); patella (TDL = 23 cm); and carpus (TDL = 27.2 cm). Secondary ossification centers first appeared in the femoral distal epiphysis (TDL = 16.6 cm) and tibial proximal epiphysis (TDL = 18.4 cm). Advanced fetuses (TDL > 30 cm, 97% gestational period) presented mineralization in all primary and most secondary centers. Compared to other species, paca neonates have a well-developed skeletal system at birth, which is important for their independent postnatal locomotion. Our results may contribute to the monitoring of bone development in other wild species, helping us to understand their life history, and serving as parameters for comparisons between precocial and altricial mammals.


Assuntos
Desenvolvimento Ósseo/fisiologia , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/embriologia , Desenvolvimento Fetal/fisiologia , Animais , Cuniculidae , Ultrassonografia
18.
Commun Biol ; 3(1): 144, 2020 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-32214226

RESUMO

The Creeper (Cp) chicken is characterized by chondrodystrophy in Cp/+ heterozygotes and embryonic lethality in Cp/Cp homozygotes. However, the genes underlying the phenotypes have not been fully known. Here, we show that a 25 kb deletion on chromosome 7, which contains the Indian hedgehog (IHH) and non-homologous end-joining factor 1 (NHEJ1) genes, is responsible for the Cp trait in Japanese bantam chickens. IHH is essential for chondrocyte maturation and is downregulated in the Cp/+ embryos and completely lost in the Cp/Cp embryos. This indicates that chondrodystrophy is caused by the loss of IHH and that chondrocyte maturation is delayed in Cp/+ heterozygotes. The Cp/Cp homozygotes exhibit impaired DNA double-strand break (DSB) repair due to the loss of NHEJ1, resulting in DSB accumulation in the vascular and nervous systems, which leads to apoptosis and early embryonic death.


Assuntos
Doenças do Desenvolvimento Ósseo/veterinária , Osso e Ossos/embriologia , Enzimas Reparadoras do DNA/genética , Proteínas de Ligação a DNA/genética , Deleção de Genes , Proteínas Hedgehog/genética , Doenças das Aves Domésticas/genética , Animais , Apoptose , Doenças do Desenvolvimento Ósseo/embriologia , Doenças do Desenvolvimento Ósseo/genética , Doenças do Desenvolvimento Ósseo/metabolismo , Osso e Ossos/metabolismo , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Embrião de Galinha , Enzimas Reparadoras do DNA/deficiência , Proteínas de Ligação a DNA/deficiência , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Predisposição Genética para Doença , Proteínas Hedgehog/deficiência , Heterozigoto , Homozigoto , Fenótipo , Doenças das Aves Domésticas/embriologia , Doenças das Aves Domésticas/metabolismo
19.
Proc Natl Acad Sci U S A ; 117(13): 7296-7304, 2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-32170021

RESUMO

Hox genes are indispensable for the proper patterning of the skeletal morphology of the axial and appendicular skeleton during embryonic development. Recently, it has been demonstrated that Hox expression continues from embryonic stages through postnatal and adult stages exclusively in a skeletal stem cell population. However, whether Hox genes continue to function after development has not been rigorously investigated. We generated a Hoxd11 conditional allele and induced genetic deletion at adult stages to show that Hox11 genes play critical roles in skeletal homeostasis of the forelimb zeugopod (radius and ulna). Conditional loss of Hox11 function at adult stages leads to replacement of normal lamellar bone with an abnormal woven bone-like matrix of highly disorganized collagen fibers. Examining the lineage from the Hox-expressing mutant cells demonstrates no loss of stem cell population. Differentiation in the osteoblast lineage initiates with Runx2 expression, which is observed similarly in mutants and controls. With loss of Hox11 function, however, osteoblasts fail to mature, with no progression to osteopontin or osteocalcin expression. Osteocyte-like cells become embedded within the abnormal bony matrix, but they completely lack dendrites, as well as the characteristic lacuno-canalicular network, and do not express SOST. Together, our studies show that Hox11 genes continuously function in the adult skeleton in a region-specific manner by regulating differentiation of Hox-expressing skeletal stem cells into the osteolineage.


Assuntos
Osso e Ossos/embriologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Animais , Osso e Ossos/metabolismo , Diferenciação Celular , Condrócitos/metabolismo , Feminino , Membro Anterior/embriologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Genes Homeobox/genética , Genes Homeobox/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/metabolismo , Esqueleto/embriologia , Fatores de Transcrição/metabolismo
20.
Development ; 147(21)2020 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-32188632

RESUMO

Bones do not normally have lymphatics. However, individuals with generalized lymphatic anomaly (GLA) or Gorham-Stout disease (GSD) develop ectopic lymphatics in bone. Despite growing interest in the development of tissue-specific lymphatics, the cellular origin of bone lymphatic endothelial cells (bLECs) is not known and the development of bone lymphatics has not been fully characterized. Here, we describe the development of bone lymphatics in mouse models of GLA and GSD. Through lineage-tracing experiments, we show that bLECs arise from pre-existing Prox1-positive LECs. We show that bone lymphatics develop in a stepwise manner where regional lymphatics grow, breach the periosteum and then invade bone. We also show that the development of bone lymphatics is impaired in mice that lack osteoclasts. Last, we show that rapamycin can suppress the growth of bone lymphatics in our models of GLA and GSD. In summary, we show that bLECs can arise from pre-existing LECs and that rapamycin can prevent the growth of bone lymphatics.


Assuntos
Osso e Ossos/embriologia , Vasos Linfáticos/embriologia , Animais , Osso e Ossos/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Integrases/metabolismo , Vasos Linfáticos/efeitos dos fármacos , Camundongos Transgênicos , Mutação/genética , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Sirolimo/farmacologia , Fator de Transcrição Sp7/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...