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










Base de datos
Intervalo de año de publicación
1.
J Bone Miner Res ; 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38696703

RESUMEN

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.

2.
J Bone Miner Res ; 38(10): 1497-1508, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37222072

RESUMEN

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).

3.
Cell Rep ; 36(8): 109618, 2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34433017

RESUMEN

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.


Asunto(s)
Médula Ósea/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Estrés Oxidativo/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Células de la Médula Ósea/citología , Movimiento Celular/fisiología , Células Cultivadas , Ratones , Nicho de Células Madre/fisiología , Trasplante de Células Madre/métodos
5.
Dev Cell ; 51(2): 236-254.e12, 2019 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-31543445

RESUMEN

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.


Asunto(s)
Regeneración Ósea/fisiología , Diferenciación Celular/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células Madre/metabolismo , Animales , Ratones , Osteogénesis/fisiología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/fisiología
7.
Int J Mol Sci ; 20(5)2019 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-30841538

RESUMEN

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.


Asunto(s)
Movimiento Celular , Distrofia Muscular de Cinturas/metabolismo , Mioblastos/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Animales , Células Cultivadas , Quimiocinas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Mioblastos/fisiología , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Sarcoglicanos/genética , Sarcoglicanos/metabolismo
8.
J Clin Invest ; 128(3): 1087-1105, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29431735

RESUMEN

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.


Asunto(s)
Glucosa/metabolismo , Osteoblastos/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Adenocarcinoma/patología , Animales , Médula Ósea/metabolismo , Neoplasias Óseas/secundario , Linaje de la Célula , Femenino , Glucólisis , Humanos , Hipoxia , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Noqueados , Mutación , Metástasis de la Neoplasia/patología , Osteocalcina/metabolismo , Transducción de Señal , Microtomografía por Rayos X
9.
J Bone Miner Res ; 32(10): 2087-2102, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28574598

RESUMEN

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.


Asunto(s)
Huesos/citología , Citoesqueleto/metabolismo , Osteogénesis , Proteínas Serina-Treonina Quinasas/metabolismo , Células Madre/citología , Vía de Señalización Wnt , Animales , Animales Recién Nacidos , Enfermedades Óseas Metabólicas/enzimología , Enfermedades Óseas Metabólicas/patología , Proteínas Morfogenéticas Óseas/metabolismo , Calcificación Fisiológica , Hueso Esponjoso/patología , Linaje de la Célula , Desarrollo Embrionario , Activación Enzimática , Femenino , Feto/embriología , Eliminación de Gen , Ratones Noqueados , Osteoblastos/enzimología , Osteoblastos/patología , Proteínas Serina-Treonina Quinasas/deficiencia , Factor de Transcripción Sp7/metabolismo , Células Madre/metabolismo
10.
Semin Cell Dev Biol ; 62: 16-33, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27180955

RESUMEN

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.


Asunto(s)
Cartílago/metabolismo , Cartílago/patología , Biología Evolutiva , Transducción de Señal , Animales , Condrocitos/metabolismo , Condrocitos/patología , Humanos , Neovascularización Fisiológica , Osteoblastos/patología
11.
12.
Stem Cells ; 32(11): 3012-22, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25069965

RESUMEN

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.


Asunto(s)
Células de la Médula Ósea/metabolismo , Médula Ósea/metabolismo , Quimiocina CXCL12/metabolismo , Células Madre Hematopoyéticas/metabolismo , Transducción de Señal , Proteínas Smad/metabolismo , Nicho de Células Madre , Animales , Proteína Morfogenética Ósea 4/metabolismo , Linaje de la Célula , Movimiento Celular/fisiología , Células Cultivadas , Regulación de la Expresión Génica/fisiología , Trasplante de Células Madre Hematopoyéticas/métodos , Ratones , Receptores CXCR4/metabolismo
13.
Birth Defects Res C Embryo Today ; 99(3): 170-91, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24078495

RESUMEN

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.


Asunto(s)
Huesos/fisiología , Homeostasis/fisiología , Osteoblastos/metabolismo , Osteogénesis/fisiología , Regeneración/fisiología , Animales , Cartílago , Diferenciación Celular , Linaje de la Célula , Humanos , Factor 1 Inducible por Hipoxia/genética , Factor 1 Inducible por Hipoxia/metabolismo , Modelos Animales , Osteoblastos/citología , Ingeniería de Tejidos , Factor A de Crecimiento Endotelial Vascular/metabolismo
14.
Calcif Tissue Int ; 92(4): 307-23, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23292135

RESUMEN

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.


Asunto(s)
Huesos/irrigación sanguínea , Huesos/fisiología , Neovascularización Fisiológica/fisiología , Animales , Desarrollo Óseo/fisiología , Homeostasis/fisiología , Humanos , Osteogénesis/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología
15.
Nat Rev Rheumatol ; 8(6): 358-66, 2012 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-22450551

RESUMEN

Adaptation to hypoxia is a critical cellular event both in pathological settings, such as cancer and ischaemia, and in normal development and differentiation. Oxygen is thought to be not only an indispensable metabolic substrate for a variety of in vivo enzymatic reactions, including mitochondrial respiration, but also a key regulatory signal in tissue development and homeostasis by controlling a specific genetic program. Hypoxia-inducible transcription factors (HIFs) HIF-1 and HIF-2 are central mediators of the homeostatic response that enables cells to survive and differentiate in low-oxygen conditions. Genetically altered mice have been used to identify important roles for HIF-1 and HIF-2 as well as vascular endothelial growth factor (VEGF)-a potent angiogenic factor and a downstream target of the HIF pathway-in the regulation of skeletal development, bone homeostasis and haematopoiesis. In this Review, we summarize the current knowledge of HIF signalling in cartilage, bone and blood, and pay particular attention to the complex relationship between HIF and VEGF in these tissues revealed by data from research using animal models. The study of these models expands our understanding of the cell autonomous, paracrine and autocrine effects that mediate the homeostatic responses downstream of HIFs and VEGF.


Asunto(s)
Desarrollo Óseo/fisiología , Enfermedades Óseas/metabolismo , Hipoxia de la Célula/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Regeneración/fisiología , Factores de Transcripción/metabolismo , Animales , Modelos Animales de Enfermedad , Homeostasis/fisiología , Humanos , Ratones , Ratones Noqueados , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
J Bone Miner Res ; 27(3): 596-609, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22162090

RESUMEN

Fetal growth plate cartilage is nonvascularized, and chondrocytes largely develop in hypoxic conditions. We previously found that mice lacking the hypoxia-inducible transcription factor HIF-1α in cartilage show massive death of centrally located, hypoxic chondrocytes. A similar phenotype was observed in mice with genetic ablation of either all or specifically the diffusible isoforms of vascular endothelial growth factor (VEGF), a prime angiogenic target of HIF-1α. Here, we assessed whether VEGF is a critical downstream component of the HIF-1α-dependent survival pathway in chondrocytes. We used a genetic approach to conditionally overexpress VEGF164 in chondrocytes lacking HIF-1α, evaluating potential rescuing effects. The effectiveness of the strategy was validated by showing that transgenic expression of VEGF164 in Col2-Cre;VEGF(f/f) mice stimulated angiogenesis in the perichondrium, fully corrected the excessive hypoxia of VEGF-deficient chondrocytes, and completely prevented chondrocyte death. Yet, similarly crossed double-mutant embryos lacking HIF-1α and overexpressing VEGF164 in the growth plate cartilage still displayed a central cell death phenotype, albeit slightly delayed and less severe compared with mice exclusively lacking HIF-1α. Transgenic VEGF164 induced massive angiogenesis in the perichondrium, yet this only partially relieved the aberrant hypoxia present in HIF-1α-deficient cartilage and thereby likely inflicted only a partial rescue effect. In fact, excessive hypoxia and failure to upregulate phosphoglycerate-kinase 1 (PGK1), a key enzyme of anaerobic glycolytic metabolism, were among the earliest manifestations of HIF-1α deficiency in cartilaginous bone templates, and reduced PGK1 expression was irrespective of transgenic VEGF164. These findings suggest that HIF-1α activates VEGF-independent cell-autonomous mechanisms to sustain oxygen levels in the challenged avascular cartilage by reducing oxygen consumption. Hence, regulation of the metabolic pathways by HIF-1α and VEGF-dependent regulation of angiogenesis coordinately act to maintain physiological cartilage oxygenation. We conclude that VEGF and HIF-1α are critical preservers of chondrocyte survival by ensuring an adequate balance between availability and handling of oxygen in developing growth cartilage.


Asunto(s)
Cartílago/fisiología , Supervivencia Celular/fisiología , Condrocitos/citología , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Consumo de Oxígeno/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Apoptosis , Cartílago/citología , Cartílago/embriología , Ratones , Ratones Transgénicos , Factor A de Crecimiento Endotelial Vascular/genética
17.
Cancer Cell ; 19(6): 740-53, 2011 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-21665148

RESUMEN

Imatinib has revolutionized the treatment of Bcr-Abl1(+) chronic myeloid leukemia (CML), but, in most patients, some leukemia cells persist despite continued therapy, while others become resistant. Here, we report that PlGF levels are elevated in CML and that PlGF produced by bone marrow stromal cells (BMSCs) aggravates disease severity. CML cells foster a soil for their own growth by inducing BMSCs to upregulate PlGF, which not only stimulates BM angiogenesis, but also promotes CML proliferation and metabolism, in part independently of Bcr-Abl1 signaling. Anti-PlGF treatment prolongs survival of imatinib-sensitive and -resistant CML mice and adds to the anti-CML activity of imatinib. These results may warrant further investigation of the therapeutic potential of PlGF inhibition for (imatinib-resistant) CML.


Asunto(s)
Proteínas de Fusión bcr-abl/fisiología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Piperazinas/uso terapéutico , Proteínas Gestacionales/fisiología , Pirimidinas/uso terapéutico , Animales , Benzamidas , Células de la Médula Ósea/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos , Humanos , Mesilato de Imatinib , Leucemia Mielógena Crónica BCR-ABL Positiva/etiología , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/mortalidad , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , FN-kappa B/fisiología , Osteólisis/prevención & control , Factor de Crecimiento Placentario , Proteínas Gestacionales/antagonistas & inhibidores , Proteínas Gestacionales/sangre
18.
Dev Cell ; 19(2): 329-44, 2010 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-20708594

RESUMEN

During endochondral bone development, the first osteoblasts differentiate in the perichondrium surrounding avascular cartilaginous rudiments; the source of trabecular osteoblasts inside the later bone is, however, unknown. Here, we generated tamoxifen-inducible transgenic mice bred to Rosa26R-LacZ reporter mice to follow the fates of stage-selective subsets of osteoblast lineage cells. Pulse-chase studies showed that osterix-expressing osteoblast precursors, labeled in the perichondrium prior to vascular invasion of the cartilage, give rise to trabecular osteoblasts, osteocytes, and stromal cells inside the developing bone. Throughout the translocation, some precursors were found to intimately associate with invading blood vessels, in pericyte-like fashion. A similar coinvasion occurs during endochondral healing of bone fractures. In contrast, perichondrial mature osteoblasts did not exhibit perivascular localization and remained in the outer cortex of developing bones. These findings reveal the specific involvement of immature osteoblast precursors in the coupled vascular and osteogenic transformation essential to endochondral bone development and repair.


Asunto(s)
Vasos Sanguíneos/metabolismo , Desarrollo Óseo/fisiología , Regeneración Ósea/fisiología , Osteoblastos/fisiología , Células Madre/fisiología , Animales , Biomarcadores/metabolismo , Vasos Sanguíneos/citología , Huesos/irrigación sanguínea , Huesos/patología , Huesos/fisiología , Linaje de la Célula , Movimiento Celular , Condrocitos/citología , Condrocitos/fisiología , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Femenino , Fracturas Óseas , Masculino , Ratones , Ratones Transgénicos , Osteoblastos/citología , Pericitos/metabolismo , Embarazo , Células Madre/citología
19.
Cancer Res ; 70(16): 6537-47, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20682798

RESUMEN

Treatment of bone metastases is largely symptomatic and is still an unmet medical need. Current therapies mainly target the late phase of tumor-induced osteoclast activation and hereby inhibit further metastatic growth. This treatment method is, however, less effective in preventing initial tumor engraftment, a process that is supposed to depend on the bone microenvironment. We explored whether bone-derived placental growth factor (PlGF), a homologue of vascular endothelial growth factor-A, regulates osteolytic metastasis. Osteogenic cells secrete PlGF, the expression of which is enhanced by bone-metastasizing breast tumor cells. Selective neutralization of host-derived PlGF by anti-mouse PlGF (alphaPlGF) reduced the incidence, number, and size of bone metastases, and preserved bone mass. alphaPlGF did not affect metastatic tumor angiogenesis but inhibited osteoclast formation by preventing the upregulation of the osteoclastogenic cytokine receptor activator of NF-kappaB ligand in osteogenic cells, as well as by blocking the autocrine osteoclastogenic activity of PlGF. alphaPlGF also reduced the engraftment of tumor cells in the bone and inhibited their interaction with matrix components in the metastatic niche. alphaPlGF therefore inhibits not only the progression of metastasis but also the settlement of tumor in the bone. These findings identify novel properties of PlGF and suggest that alphaPlGF might offer opportunities for adjuvant therapy of bone metastasis.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias de la Mama/patología , Osteoclastos/patología , Proteínas Gestacionales/antagonistas & inhibidores , Proteínas Gestacionales/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Neoplasias Óseas/metabolismo , Neoplasias de la Mama/metabolismo , Diferenciación Celular/fisiología , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Pulmonares/secundario , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Osteoclastos/metabolismo , Factor de Crecimiento Placentario , Trasplante Heterólogo
20.
EMBO J ; 29(2): 424-41, 2010 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-20010698

RESUMEN

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.


Asunto(s)
Huesos/metabolismo , Huesos/patología , Regulación del Desarrollo de la Expresión Génica , Factor A de Crecimiento Endotelial Vascular/metabolismo , beta Catenina/metabolismo , Animales , Huesos/embriología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Células Endoteliales/citología , Humanos , Mesodermo/citología , Ratones , Ratones Transgénicos , Morfogénesis , Osteoblastos/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Células Madre/citología , Células del Estroma/citología , Factor A de Crecimiento Endotelial Vascular/genética , beta Catenina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...