Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Semin Cell Dev Biol ; 62: 16-33, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27180955

RESUMO

Endochondral ossification, the mechanism responsible for the development of the long bones, is dependent on an extremely stringent coordination between the processes of chondrocyte maturation in the growth plate, vascular expansion in the surrounding tissues, and osteoblast differentiation and osteogenesis in the perichondrium and the developing bone center. The synchronization of these processes occurring in adjacent tissues is regulated through vigorous crosstalk between chondrocytes, endothelial cells and osteoblast lineage cells. Our knowledge about the molecular constituents of these bidirectional communications is undoubtedly incomplete, but certainly some signaling pathways effective in cartilage have been recognized to play key roles in steering vascularization and osteogenesis in the perichondrial tissues. These include hypoxia-driven signaling pathways, governed by the hypoxia-inducible factors (HIFs) and vascular endothelial growth factor (VEGF), which are absolutely essential for the survival and functioning of chondrocytes in the avascular growth plate, at least in part by regulating the oxygenation of developing cartilage through the stimulation of angiogenesis in the surrounding tissues. A second coordinating signal emanating from cartilage and regulating developmental processes in the adjacent perichondrium is Indian Hedgehog (IHH). IHH, produced by pre-hypertrophic and early hypertrophic chondrocytes in the growth plate, induces the differentiation of adjacent perichondrial progenitor cells into osteoblasts, thereby harmonizing the site and time of bone formation with the developmental progression of chondrogenesis. Both signaling pathways represent vital mediators of the tightly organized conversion of avascular cartilage into vascularized and mineralized bone during endochondral ossification.


Assuntos
Cartilagem/metabolismo , Cartilagem/patologia , Biologia do Desenvolvimento , Transdução de Sinais , Animais , Condrócitos/metabolismo , Condrócitos/patologia , Humanos , Neovascularização Fisiológica , Osteoblastos/patologia
2.
Int J Mol Sci ; 20(5)2019 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-30841538

RESUMO

Progressive muscle degeneration followed by dilated cardiomyopathy is a hallmark of muscular dystrophy. Stem cell therapy is suggested to replace diseased myofibers by healthy myofibers, although so far, we are faced by low efficiencies of migration and engraftment of stem cells. Chemokines are signalling proteins guiding cell migration and have been shown to tightly regulate muscle tissue repair. We sought to determine which chemokines are expressed in dystrophic muscles undergoing tissue remodelling. Therefore, we analysed the expression of chemokines and chemokine receptors in skeletal and cardiac muscles from Sarcoglycan-α null, Sarcoglycan-ß null and immunodeficient Sgcß-null mice. We found that several chemokines are dysregulated in dystrophic muscles. We further show that one of these, platelet-derived growth factor-B, promotes interstitial stem cell migration. This finding provides perspective to an approachable mechanism for improving stem cell homing towards dystrophic muscles.


Assuntos
Movimento Celular , Distrofia Muscular do Cíngulo dos Membros/metabolismo , Mioblastos/metabolismo , Proteínas Proto-Oncogênicas c-sis/metabolismo , Animais , Células Cultivadas , Quimiocinas/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mioblastos/fisiologia , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Sarcoglicanas/genética , Sarcoglicanas/metabolismo
3.
Stem Cells ; 32(11): 3012-22, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25069965

RESUMO

We recently demonstrated that ex vivo activation of SMAD-independent bone morphogenetic protein 4 (BMP4) signaling in hematopoietic stem/progenitor cells (HSPCs) influences their homing into the bone marrow (BM). Here, we assessed whether alterations in BMP signaling in vivo affects adult hematopoiesis by affecting the BM niche. We demonstrate that systemic inhibition of SMAD-dependent BMP signaling by infusion of the BMP antagonist noggin (NGN) significantly increased CXCL12 levels in BM plasma leading to enhanced homing and engraftment of transplanted HSPCs. Conversely, the infusion of BMP7 but not BMP4, resulted in decreased HSPC homing. Using ST2 cells as an in vitro model of BM niche, we found that incubation with neutralizing anti-BMP4 antibodies, NGN, or dorsomorphin (DM) as well as knockdown of Smad1/5 and Bmp4, all enhanced CXCL12 production. Chromatin immunoprecipitation identified the SMAD-binding element in the CXCL12 promoter to which SMAD4 binds. When deleted, increased CXCL12 promoter activity was observed, and NGN or DM no longer affected Cxcl12 expression. Interestingly, BMP7 infusion resulted in mobilization of only short-term HSCs, likely because BMP7 affected CXCL12 expression only in osteoblasts but not in other niche components. Hence, we describe SMAD-dependent BMP signaling as a novel regulator of CXCL12 production in the BM niche, influencing HSPC homing, engraftment, and mobilization.


Assuntos
Células da Medula Óssea/metabolismo , Medula Óssea/metabolismo , Quimiocina CXCL12/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo , Nicho de Células-Tronco , Animais , Proteína Morfogenética Óssea 4/metabolismo , Linhagem da Célula , Movimento Celular/fisiologia , Células Cultivadas , Regulação da Expressão Gênica/fisiologia , Transplante de Células-Tronco Hematopoéticas/métodos , Camundongos , Receptores CXCR4/metabolismo
4.
EMBO J ; 29(2): 424-41, 2010 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-20010698

RESUMO

Vascular endothelial growth factor (VEGF) and beta-catenin both act broadly in embryogenesis and adulthood, including in the skeletal and vascular systems. Increased or deregulated activity of these molecules has been linked to cancer and bone-related pathologies. By using novel mouse models to locally increase VEGF levels in the skeleton, we found that embryonic VEGF over-expression in osteo-chondroprogenitors and their progeny largely pheno-copied constitutive beta-catenin activation. Adult induction of VEGF in these cell populations dramatically increased bone mass, associated with aberrant vascularization, bone marrow fibrosis and haematological anomalies. Genetic and pharmacological interventions showed that VEGF increased bone mass through a VEGF receptor 2- and phosphatidyl inositol 3-kinase-mediated pathway inducing beta-catenin transcriptional activity in endothelial and osteoblastic cells, likely through modulation of glycogen synthase kinase 3-beta phosphorylation. These insights into the actions of VEGF in the bone and marrow environment underscore its power as pleiotropic bone anabolic agent but also warn for caution in its therapeutic use. Moreover, the finding that VEGF can modulate beta-catenin activity may have widespread physiological and clinical ramifications.


Assuntos
Osso e Ossos/metabolismo , Osso e Ossos/patologia , Regulação da Expressão Gênica no Desenvolvimento , Fator A de Crescimento do Endotélio Vascular/metabolismo , beta Catenina/metabolismo , Animais , Osso e Ossos/embriologia , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Células Endoteliais/citologia , Humanos , Mesoderma/citologia , Camundongos , Camundongos Transgênicos , Morfogênese , Osteoblastos/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Células-Tronco/citologia , Células Estromais/citologia , Fator A de Crescimento do Endotélio Vascular/genética , beta Catenina/genética
5.
J Bone Miner Res ; 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38696703

RESUMO

Bone development, growth, and repair are complex processes involving various cell types and interactions, with central roles played by skeletal stem and progenitor cells. Recent research brought new insights into the skeletal precursor populations that mediate intramembranous and endochondral bone development. Later in life, many of the cellular and molecular mechanisms determining development are reactivated upon fracture, with powerful trauma-induced signaling cues triggering a variety of postnatal skeletal stem/progenitor cells (SSPCs) residing near the bone defect. Interestingly, in this injury context, the current evidence suggests that the fates of both SSPCs and differentiated skeletal cells can be considerably flexible and dynamic, and that multiple cell sources can be activated to operate as functional progenitors generating chondrocytes and/or osteoblasts. The combined implementation of in vivo lineage tracing, cell surface marker-based selection, single-cell molecular analyses, and high-resolution in situ imaging has strongly improved our insights into the diversity and roles of developmental and reparative stem/progenitor subsets, while also unveiling the complexity of their dynamics, hierarchies, and relationships. Albeit incompletely understood at present, findings supporting lineage flexibility and possibly plasticity among sources of osteogenic cells challenge the classical dogma of a single primitive, self-renewing, multipotent stem cell driving bone tissue formation and regeneration from the apex of a hierarchical and strictly unidirectional differentiation tree. We here review the state of the field and the newest discoveries in the origin, identity, and fates of skeletal progenitor cells during bone development and growth, discuss the contributions of adult SSPC populations to fracture repair, and reflect on the dynamism and relationships among skeletal precursors and differentiated cell lineages. Further research directed at unraveling the heterogeneity and capacities of SSPCs, as well as the regulatory cues determining their fate and functioning, will offer vital new options for clinical translation toward compromised fracture healing and bone regenerative medicine.


Skeletal progenitor cells are crucial for bone development and growth, as they provide the cellular building blocks (chondrocytes and osteoblasts) that form the cartilage and bone tissues that the skeleton is composed of. In adult life, the occurrence of a bone fracture reactivates similar tissue-forming mechanisms, starting with the trauma triggering various postnatal skeletal stem/progenitor cells (SSPCs) residing near the bone defect to divide and migrate. These cells subsequently generate functional fracture-repairing cells by differentiating into mature chondrocytes and/or osteoblasts. In recent years, the combined use of various advanced research approaches and new techniques has strongly improved our insights into the origin, identity, fates, and roles of developmental and reparative skeletal stem cells and progenitor subsets. Concomitantly, this research also unveiled considerable complexity in their dynamics, diversity, hierarchies, and relationships, which is incompletely understood at present. In this review, we discuss the state of the field and the newest discoveries in the identity and roles of skeletal stem and progenitor cells mediating bone development, growth, and repair. Further research on these cell populations, including determining their exact nature, fate, and functioning, and how they can be harvested and regulated, is critical to develop new treatments for non-healing fractures.

6.
Birth Defects Res C Embryo Today ; 99(3): 170-91, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24078495

RESUMO

During endochondral bone development, bone-forming osteoblasts have to colonize the regions of cartilage that will be replaced by bone. In adulthood, bone remodeling and repair require osteogenic cells to reach the sites that need to be rebuilt, as a prerequisite for skeletal health. A failure of osteoblasts to reach the sites in need of bone formation may contribute to impaired fracture repair. Conversely, stimulation of osteogenic cell recruitment may be a promising osteo-anabolic strategy to improve bone formation in low bone mass disorders such as osteoporosis and in bone regeneration applications. Yet, still relatively little is known about the cellular and molecular mechanisms controlling osteogenic cell recruitment to sites of bone formation. In vitro, several secreted growth factors have been shown to induce osteogenic cell migration. Recent studies have started to shed light on the role of such chemotactic signals in the regulation of osteoblast recruitment during bone remodeling. Moreover, trafficking of osteogenic cells during endochondral bone development and repair was visualized in vivo by lineage tracing, revealing that the capacity of osteoblast lineage cells to move into new bone centers is largely confined to undifferentiated osteoprogenitors, and coupled to angiogenic invasion of the bone-modeling cartilage intermediate. It is well known that the presence of blood vessels is absolutely required for bone formation, and that a close spatial and temporal relationship exists between osteogenesis and angiogenesis. Studies using genetically modified mouse models have identified some of the molecular constituents of this osteogenic-angiogenic coupling. This article reviews the current knowledge on the process of osteoblast lineage cell recruitment to sites of active bone formation in skeletal development, remodeling, and repair, considering the role of chemo-attractants for osteogenic cells and the interplay between osteogenesis and angiogenesis in the control of bone formation.


Assuntos
Osso e Ossos/fisiologia , Homeostase/fisiologia , Osteoblastos/metabolismo , Osteogênese/fisiologia , Regeneração/fisiologia , Animais , Cartilagem , Diferenciação Celular , Linhagem da Célula , Humanos , Fator 1 Induzível por Hipóxia/genética , Fator 1 Induzível por Hipóxia/metabolismo , Modelos Animais , Osteoblastos/citologia , Engenharia Tecidual , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Calcif Tissue Int ; 92(4): 307-23, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23292135

RESUMO

Adequate vascularization is an absolute requirement for bone development, growth, homeostasis, and repair. Endochondral ossification during fetal skeletogenesis is typified by the initial formation of a prefiguring cartilage template of the future bone, which itself is intrinsically avascular. When the chondrocytes reach terminal hypertrophic differentiation they become invaded by blood vessels. This neovascularization process triggers the progressive replacement of the growing cartilage by bone, in a complex multistep process that involves the coordinated activity of chondrocytes, osteoblasts, and osteoclasts, each standing in functional interaction with the vascular system. Studies using genetically modified mice have started to shed light on the molecular regulation of the cartilage neovascularization processes that drive endochondral bone development, growth, and repair, with a prime role being played by vascular endothelial growth factor and its isoforms. The vasculature of bone remains important throughout life as an intrinsic component of the bone and marrow environment. Bone remodeling, the continual renewal of bone by the balanced activities of osteoclasts resorbing packets of bone and osteoblasts building new bone, takes place in close spatial relationship with the vascular system and depends on signals, oxygen, and cellular delivery via the bloodstream. Conversely, the integrity and functionality of the vessel system, including the exchange of blood cells between the hematopoietic marrow and the circulation, rely on a delicate interplay with the cells of bone. Here, the current knowledge on the cellular relationships and molecular crosstalk that coordinate skeletal vascularization in bone development and homeostasis will be reviewed.


Assuntos
Osso e Ossos/irrigação sanguínea , Osso e Ossos/fisiologia , Neovascularização Fisiológica/fisiologia , Animais , Desenvolvimento Ósseo/fisiologia , Homeostase/fisiologia , Humanos , Osteogênese/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
8.
J Bone Miner Res ; 38(10): 1497-1508, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37222072

RESUMO

Transgender youth increasingly present at pediatric gender services. Some of them receive long-term puberty suppression with gonadotropin-releasing hormone analogues (GnRHa) before starting gender-affirming hormones (GAH). The impact of GnRHa use started in early puberty on bone composition and bone mass accrual is unexplored. It is furthermore unclear whether subsequent GAH fully restore GnRHa effects and whether the timing of GAH introduction matters. To answer these questions, we developed a mouse model mimicking the clinical strategy applied in trans boys. Prepubertal 4-week-old female mice were treated with GnRHa alone or with GnRHa supplemented with testosterone (T) from 6 weeks (early puberty) or 8 weeks (late puberty) onward. Outcomes were analyzed at 16 weeks and compared with untreated mice of both sexes. GnRHa markedly increased total body fat mass, decreased lean body mass, and had a modest negative impact on grip strength. Both early and late T administration shaped body composition to adult male levels, whereas grip strength was restored to female values. GnRHa-treated animals showed lower trabecular bone volume and reduced cortical bone mass and strength. These changes were reversed by T to female levels (cortical bone mass and strength) irrespective of the time of administration or even fully up to adult male control values (trabecular parameters) in case of earlier T start. The lower bone mass in GnRHa-treated mice was associated with increased bone marrow adiposity, also reversed by T. In conclusion, prolonged GnRHa use started in prepubertal female mice modifies body composition toward more fat and less lean mass and impairs bone mass acquisition and strength. Subsequent T administration counteracts GnRHa impact on these parameters, shaping body composition and trabecular parameters to male values while restoring cortical bone architecture and strength up to female but not male control levels. These findings could help guide clinical strategies in transgender care. © 2023 American Society for Bone and Mineral Research (ASBMR).

9.
Nat Med ; 9(2): 173-82, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12539040

RESUMO

Hemizygous deletion of chromosome 22q11 (del22q11) causes thymic, parathyroid, craniofacial and life-threatening cardiovascular birth defects in 1 in 4,000 infants. The del22q11 syndrome is likely caused by haploinsufficiency of TBX1, but its variable expressivity indicates the involvement of additional modifiers. Here, we report that absence of the Vegf164 isoform caused birth defects in mice, reminiscent of those found in del22q11 patients. The close correlation of birth and vascular defects indicated that vascular dysgenesis may pathogenetically contribute to the birth defects. Vegf interacted with Tbx1, as Tbx1 expression was reduced in Vegf164-deficient embryos and knocked-down vegf levels enhanced the pharyngeal arch artery defects induced by tbx1 knockdown in zebrafish. Moreover, initial evidence suggested that a VEGF promoter haplotype was associated with an increased risk for cardiovascular birth defects in del22q11 individuals. These genetic data in mouse, fish and human indicate that VEGF is a modifier of cardiovascular birth defects in the del22q11 syndrome.


Assuntos
Deleção Cromossômica , Síndrome de DiGeorge/genética , Fatores de Crescimento Endotelial/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Linfocinas/genética , Animais , Vasos Sanguíneos/anormalidades , Anormalidades Congênitas/genética , Face/anormalidades , Camundongos , Camundongos Knockout , Neuropilina-1/genética , Isoformas de Proteínas/genética , Crânio/anormalidades , Proteínas com Domínio T/genética , Timo/anormalidades , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular , Peixe-Zebra
10.
Cell Rep ; 36(8): 109618, 2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34433017

RESUMO

Hematopoietic stem and progenitor cell (HSPC) engraftment after transplantation during anticancer treatment depends on support from the recipient bone marrow (BM) microenvironment. Here, by studying physiological homing of fetal HSPCs, we show the critical requirement of balanced local crosstalk within the skeletal niche for successful HSPC settlement in BM. Transgene-induced overproduction of vascular endothelial growth factor (VEGF) by osteoprogenitor cells elicits stromal and endothelial hyperactivation, profoundly impacting the stromal-vessel interface and vascular architecture. Concomitantly, HSPC homing and survival are drastically impaired. Transcriptome profiling, flow cytometry, and high-resolution imaging indicate alterations in perivascular and endothelial cell characteristics, vascular function and cellular metabolism, associated with increased oxidative stress within the VEGF-enriched BM environment. Thus, developmental HSPC homing to bone is controlled by local stromal-vascular integrity and the oxidative-metabolic status of the recipient milieu. Interestingly, irradiation of adult mice also induces stromal VEGF expression and similar osteo-angiogenic niche changes, underscoring that our findings may contribute targets for improving stem cell therapies.


Assuntos
Medula Óssea/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Estresse Oxidativo/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Células da Medula Óssea/citologia , Movimento Celular/fisiologia , Células Cultivadas , Camundongos , Nicho de Células-Tronco/fisiologia , Transplante de Células-Tronco/métodos
11.
J Clin Invest ; 116(12): 3150-9, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17099775

RESUMO

Genomic actions induced by 1alpha25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3)] are crucial for normal bone metabolism, mainly because they regulate active intestinal calcium transport. To evaluate whether the vitamin D receptor (VDR) has a specific role in growth-plate development and endochondral bone formation, we investigated mice with conditional inactivation of VDR in chondrocytes. Growth-plate chondrocyte development was not affected by the lack of VDR. Yet vascular invasion was impaired, and osteoclast number was reduced in juvenile mice, resulting in increased trabecular bone mass. In vitro experiments confirmed that VDR signaling in chondrocytes directly regulated osteoclastogenesis by inducing receptor activator of NF-kappaB ligand (RANKL) expression. Remarkably, mineral homeostasis was also affected in chondrocyte-specific VDR-null mice, as serum phosphate and 1,25(OH)(2)D levels were increased in young mice, in whom growth-plate activity is important. Both in vivo and in vitro analysis indicated that VDR inactivation in chondrocytes reduced the expression of FGF23 by osteoblasts and consequently led to increased renal expression of 1alpha-hydroxylase and of sodium phosphate cotransporter type IIa. Taken together, our findings provide evidence that VDR signaling in chondrocytes is required for timely osteoclast formation during bone development and for the endocrine action of bone in phosphate homeostasis.


Assuntos
Condrócitos/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Osteoblastos/metabolismo , Receptores de Calcitriol/fisiologia , Animais , Animais Recém-Nascidos , Desenvolvimento Ósseo/genética , Desenvolvimento Ósseo/fisiologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Condrócitos/citologia , Fator de Crescimento de Fibroblastos 23 , Expressão Gênica/genética , Lâmina de Crescimento/citologia , Lâmina de Crescimento/metabolismo , Homeostase/fisiologia , Imuno-Histoquímica , Camundongos , Mutação/genética , Osteoblastos/citologia , Osteoclastos/citologia , Osteoclastos/metabolismo , Osteogênese/genética , Osteogênese/fisiologia , Fosfatos/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Ligante RANK/genética , Ligante RANK/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Calcitriol/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fatores de Tempo , Vitamina D/análogos & derivados , Vitamina D/genética , Vitamina D/fisiologia
12.
J Clin Invest ; 116(5): 1230-42, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16614757

RESUMO

Current therapies for delayed- or nonunion bone fractures are still largely ineffective. Previous studies indicated that the VEGF homolog placental growth factor (PlGF) has a more significant role in disease than in health. Therefore we investigated the role of PlGF in a model of semi-stabilized bone fracture healing. Fracture repair in mice lacking PlGF was impaired and characterized by a massive accumulation of cartilage in the callus, reminiscent of delayed- or nonunion fractures. PlGF was required for the early recruitment of inflammatory cells and the vascularization of the fracture wound. Interestingly, however, PlGF also played a role in the subsequent stages of the repair process. Indeed in vivo and in vitro findings indicated that PlGF induced the proliferation and osteogenic differentiation of mesenchymal progenitors and stimulated cartilage turnover by particular MMPs. Later in the process, PlGF was required for the remodeling of the newly formed bone by stimulating osteoclast differentiation. As PlGF expression was increased throughout the process of bone repair and all the important cell types involved expressed its receptor VEGFR-1, the present data suggest that PlGF is required for mediating and coordinating the key aspects of fracture repair. Therefore PlGF may potentially offer therapeutic advantages for fracture repair.


Assuntos
Remodelação Óssea , Cartilagem/citologia , Consolidação da Fratura , Mesoderma/citologia , Proteínas da Gravidez/fisiologia , Animais , Cartilagem/metabolismo , Diferenciação Celular , Proliferação de Células , Inflamação , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Osteoclastos/citologia , Fator de Crescimento Placentário , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
13.
Dev Cell ; 51(2): 236-254.e12, 2019 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-31543445

RESUMO

Bone repair and regeneration critically depend on the activation and recruitment of osteogenesis-competent skeletal stem and progenitor cells (SSPCs). Yet, the origin and triggering cues for SSPC propagation and migration remain largely elusive. Through bulk and single-cell transcriptome profiling of fetal osterix (Osx)-expressing cells, followed by lineage mapping, cell tracing, and conditional mouse mutagenesis, we here identified PDGF-PDGFRß signaling as critical functional mediator of SSPC expansion, migration, and angiotropism during bone repair. Our data show that cells marked by a history of Osx expression, including those arising in fetal or early postnatal periods, represent or include SSPCs capable of delivering all the necessary differentiated progeny to repair acute skeletal injuries later in life, provided that they express functional PDGFRß. Mechanistically, MMP-9 and VCAM-1 appear to be involved downstream of PDGF-PDGFRß. Our results reveal considerable cellular dynamism in the skeletal system and show that activation and recruitment of SSPCs for bone repair require functional PDGFRß signaling.


Assuntos
Regeneração Óssea/fisiologia , Diferenciação Celular/fisiologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Células-Tronco/metabolismo , Animais , Camundongos , Osteogênese/fisiologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais/fisiologia
14.
J Clin Invest ; 128(3): 1087-1105, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29431735

RESUMO

The skeleton has emerged as an important regulator of systemic glucose homeostasis, with osteocalcin and insulin representing prime mediators of the interplay between bone and energy metabolism. However, genetic evidence indicates that osteoblasts can influence global energy metabolism through additional, as yet unknown, mechanisms. Here, we report that constitutive or postnatally induced deletion of the hypoxia signaling pathway component von Hippel-Lindau (VHL) in skeletal osteolineage cells of mice led to high bone mass as well as hypoglycemia and increased glucose tolerance, not accounted for by osteocalcin or insulin. In vitro and in vivo data indicated that Vhl-deficient osteoblasts displayed massively increased glucose uptake and glycolysis associated with upregulated HIF-target gene expression, resembling the Warburg effect that typifies cancer cells. Overall, the glucose consumption by the skeleton was increased in the mutant mice, as revealed by 18F-FDG radioactive tracer experiments. Moreover, the glycemia levels correlated inversely with the level of skeletal glucose uptake, and pharmacological treatment with the glycolysis inhibitor dichloroacetate (DCA), which restored glucose metabolism in Vhl-deficient osteogenic cells in vitro, prevented the development of the systemic metabolic phenotype in the mutant mice. Altogether, these findings reveal a novel link between cellular glucose metabolism in osteoblasts and whole-body glucose homeostasis, controlled by local hypoxia signaling in the skeleton.


Assuntos
Glucose/metabolismo , Osteoblastos/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Adenocarcinoma/patologia , Animais , Medula Óssea/metabolismo , Neoplasias Ósseas/secundário , Linhagem da Célula , Feminino , Glicólise , Humanos , Hipóxia , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Knockout , Mutação , Metástase Neoplásica/patologia , Osteocalcina/metabolismo , Transdução de Sinais , Microtomografia por Raio-X
15.
J Clin Invest ; 113(2): 188-99, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14722611

RESUMO

VEGF is crucial for metaphyseal bone vascularization. In contrast, the angiogenic factors required for vascularization of epiphyseal cartilage are unknown, although this represents a developmentally and clinically important aspect of bone growth. The VEGF gene is alternatively transcribed into VEGF(120), VEGF(164), and VEGF(188) isoforms that differ in matrix association and receptor binding. Their role in bone development was studied in mice expressing single isoforms. Here we report that expression of only VEGF(164) or only VEGF(188) (in VEGF(188/188) mice) was sufficient for metaphyseal development. VEGF(188/188) mice, however, showed dwarfism, disrupted development of growth plates and secondary ossification centers, and knee joint dysplasia. This phenotype was at least partly due to impaired vascularization surrounding the epiphysis, resulting in ectopically increased hypoxia and massive chondrocyte apoptosis in the interior of the epiphyseal cartilage. In addition to the vascular defect, we provide in vitro evidence that the VEGF(188) isoform alone is also insufficient to regulate chondrocyte proliferation and survival responses to hypoxia. Consistent herewith, chondrocytes in or close to the hypoxic zone in VEGF(188/188) mice showed increased proliferation and decreased differentiation. These findings indicate that the insoluble VEGF(188) isoform is insufficient for establishing epiphyseal vascularization and regulating cartilage development during endochondral bone formation.


Assuntos
Condrócitos/citologia , Epífises/irrigação sanguínea , Epífises/metabolismo , Fator A de Crescimento do Endotélio Vascular/química , Fator A de Crescimento do Endotélio Vascular/genética , Proteínas de Xenopus , Angiografia , Animais , Desenvolvimento Ósseo , Bromodesoxiuridina/farmacologia , Cartilagem/citologia , Cartilagem/patologia , Diferenciação Celular , Divisão Celular , Sobrevivência Celular , Condrócitos/metabolismo , DNA Complementar/metabolismo , Hipóxia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Modelos Biológicos , Mutagênese , Neovascularização Fisiológica , Proteínas do Tecido Nervoso/metabolismo , Fenótipo , Ligação Proteica , Isoformas de Proteínas , RNA Mensageiro/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Recombinação Genética , Ribonucleoproteínas/metabolismo
16.
Ann N Y Acad Sci ; 1116: 149-64, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18083926

RESUMO

Over the past few decades, osteoblast differentiation has been studied extensively in a variety of culture systems and findings from these experiments have shaped our understanding of the bone-forming cell lineage. However, in vitro assays are bound by intrinsic limitations and are unable to effectively mirror many aspects related to osteoblasts in vivo, including their origin, destiny, and life span. Therefore, these fundamental questions strongly advocate the need for novel models to characterize the osteoblast lineage in vivo. Here, we developed a transgenic mouse system to study stage-specific subsets of osteoblast lineage cells. We believe that this system will prove to be a helpful tool in deciphering multiple aspects of osteoblast biology in vivo.


Assuntos
Osteoblastos/citologia , Animais , Diferenciação Celular , Linhagem da Célula , Camundongos , Camundongos Transgênicos , Modelos Animais
17.
J Bone Miner Res ; 32(10): 2087-2102, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28574598

RESUMO

Cell-matrix interactions constitute a fundamental aspect of skeletal cell biology and play essential roles in bone homeostasis. These interactions are primarily mediated by transmembrane integrin receptors, which mediate cell adhesion and transduce signals from the extracellular matrix to intracellular responses via various downstream effectors, including integrin-linked kinase (ILK). ILK functions as adaptor protein at focal adhesion sites, linking integrins to the actin cytoskeleton, and has been reported to act as a kinase phosphorylating signaling molecules such as GSK-3ß and Akt. Thereby, ILK plays important roles in cellular attachment, motility, proliferation and survival. To assess the in vivo role of ILK signaling in osteoprogenitors and the osteoblast lineage cells descending thereof, we generated conditional knockout mice using the Osx-Cre:GFP driver strain. Mice lacking functional ILK in osterix-expressing cells and their derivatives showed no apparent developmental or growth phenotype, but by 5 weeks of age they displayed a significantly reduced trabecular bone mass, which persisted into adulthood in male mice. Histomorphometry and serum analysis indicated no alterations in osteoclast formation and activity, but provided evidence that osteoblast function was impaired, resulting in reduced bone mineralization and increased accumulation of unmineralized osteoid. In vitro analyses further substantiated that absence of ILK in osteogenic cells was associated with compromised collagen matrix production and mineralization. Mechanistically, we found evidence for both impaired cytoskeletal functioning and reduced signal transduction in osteoblasts lacking ILK. Indeed, loss of ILK in primary osteogenic cells impaired F-actin organization, cellular adhesion, spreading, and migration, indicative of defective coupling of cell-matrix interactions to the cytoskeleton. In addition, BMP/Smad and Wnt/ß-catenin signaling was reduced in the absence of ILK. Taken together, these data demonstrate the importance of integrin-mediated cell-matrix interactions and ILK signaling in osteoprogenitors in the control of osteoblast functioning during juvenile bone mass acquisition and adult bone remodeling and homeostasis. © 2017 American Society for Bone and Mineral Research.


Assuntos
Osso e Ossos/citologia , Citoesqueleto/metabolismo , Osteogênese , Proteínas Serina-Treonina Quinases/metabolismo , Células-Tronco/citologia , Via de Sinalização Wnt , Animais , Animais Recém-Nascidos , Doenças Ósseas Metabólicas/enzimologia , Doenças Ósseas Metabólicas/patologia , Proteínas Morfogenéticas Ósseas/metabolismo , Calcificação Fisiológica , Osso Esponjoso/patologia , Linhagem da Célula , Desenvolvimento Embrionário , Ativação Enzimática , Feminino , Feto/embriologia , Deleção de Genes , Camundongos Knockout , Osteoblastos/enzimologia , Osteoblastos/patologia , Proteínas Serina-Treonina Quinases/deficiência , Fator de Transcrição Sp7/metabolismo , Células-Tronco/metabolismo
18.
Med Biol Eng Comput ; 44(4): 280-9, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16937169

RESUMO

The combined use of experimental and mathematical models can lead to a better understanding of fracture healing. In this study, a mathematical model, which was originally established by Bailón-Plaza and van der Meulen (J Theor Biol 212:191-209, 2001), was applied to an experimental model of a semi-stabilized murine tibial fracture. The mathematical model was implemented in a custom finite volumes code, specialized in dealing with the model's requirements of mass conservation and non-negativity of the variables. A qualitative agreement between the experimentally measured and numerically simulated evolution in the cartilage and bone content was observed. Additionally, an extensive parametric study was conducted to assess the influence of the model parameters on the simulation outcome. Finally, a case of pathological fracture healing and its treatment by administration of growth factors was modeled to demonstrate the potential therapeutic value of this mathematical model.


Assuntos
Simulação por Computador , Consolidação da Fratura , Animais , Cartilagem/diagnóstico por imagem , Cartilagem/lesões , Camundongos , Modelos Animais , Radiografia , Fraturas da Tíbia/diagnóstico por imagem
19.
Mech Dev ; 111(1-2): 61-73, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11804779

RESUMO

Vascular endothelial growth factor (VEGF)-mediated angiogenesis is an important part of bone formation. To clarify the role of VEGF isoforms in endochondral bone formation, we examined long bone development in mice expressing exclusively the VEGF120 isoform (VEGF120/120 mice). Neonatal VEGF120/120 long bones showed a completely disturbed vascular pattern, concomitant with a 35% decrease in trabecular bone volume, reduced bone growth and a 34% enlargement of the hypertrophic chondrocyte zone of the growth plate. Surprisingly, embryonic hindlimbs at a stage preceding capillary invasion exhibited a delay in bone collar formation and hypertrophic cartilage calcification. Expression levels of marker genes of osteoblast and hypertrophic chondrocyte differentiation were significantly decreased in VEGF120/120 bones. Furthermore, inhibition of all VEGF isoforms in cultures of embryonic cartilaginous metatarsals, through the administration of a soluble receptor chimeric protein (mFlt-1/Fc), retarded the onset and progression of ossification, suggesting that osteoblast and/or hypertrophic chondrocyte development were impaired. The initial invasion by osteoclasts and endothelial cells into VEGF120/120 bones was retarded, associated with decreased expression of matrix metalloproteinase-9. Our findings indicate that expression of VEGF164 and/or VEGF188 is important for normal endochondral bone development, not only to mediate bone vascularization but also to allow normal differentiation of hypertrophic chondrocytes, osteoblasts, endothelial cells and osteoclasts.


Assuntos
Desenvolvimento Ósseo , Osso e Ossos/irrigação sanguínea , Fatores de Crescimento Endotelial/fisiologia , Linfocinas/fisiologia , Neovascularização Fisiológica/genética , Animais , Cartilagem/anormalidades , Cartilagem/embriologia , Diferenciação Celular/genética , Condrócitos/patologia , Condrócitos/fisiologia , Endotélio Vascular/patologia , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Ossos do Metatarso/embriologia , Ossos do Metatarso/patologia , Camundongos , Camundongos Mutantes , Ossificação Heterotópica , Osteoblastos/patologia , Osteoblastos/fisiologia , Osteoclastos/patologia , Osteoclastos/fisiologia , Isoformas de Proteínas/fisiologia , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA