Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Acta Astronaut ; 92(1): 73-78, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23976802

RESUMEN

Skeletal loading and unloading has a pronounced impact on bone remodeling, a process also regulated by insulin-like growth factor 1 (IGF-1) signaling. Skeletal unloading leads to resistance to the anabolic effect of IGF-1, while reloading after unloading restores responsiveness to IGF-1. However, a direct study of the importance of IGF-1 signaling in the skeletal response to mechanical loading remains to be tested. In this study, we assessed the skeletal response of osteoblast-specific Igf-1 receptor deficient (Igf-1r-/- ) mice to unloading and reloading. The mice were hindlimb unloaded for 14 days and then reloaded for 16 days. Igf-1r-/- mice displayed smaller cortical bone and diminished periosteal and endosteal bone formation at baseline. Periosteal and endosteal bone formation decreased with unloading in Igf-1r+/+ mice. However, the recovery of periosteal bone formation with reloading was completely inhibited in Igf-1r-/- mice, although reloading-induced endosteal bone formation was not hampered. These changes in bone formation resulted in the abolishment of the expected increase in total cross-sectional area with reloading in Igf-1r-/- mice compared to the control mice. These results suggest that the Igf-1r in mature osteoblasts has a critical role in periosteal bone formation in the skeletal response to mechanical loading.

2.
J Clin Invest ; 117(3): 803-11, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17290304

RESUMEN

An essential element of the innate immune response to injury is the capacity to recognize microbial invasion and stimulate production of antimicrobial peptides. We investigated how this process is controlled in the epidermis. Keratinocytes surrounding a wound increased expression of the genes coding for the microbial pattern recognition receptors CD14 and TLR2, complementing an increase in cathelicidin antimicrobial peptide expression. These genes were induced by 1,25(OH)2 vitamin D3 (1,25D3; its active form), suggesting a role for vitamin D3 in this process. How 1,25D3 could participate in the injury response was explained by findings that the levels of CYP27B1, which converts 25OH vitamin D3 (25D3) to active 1,25D3, were increased in wounds and induced in keratinocytes in response to TGF-beta1. Blocking the vitamin D receptor, inhibiting CYP27B1, or limiting 25D3 availability prevented TGF-beta1 from inducing cathelicidin, CD14, or TLR2 in human keratinocytes, while CYP27B1-deficient mice failed to increase CD14 expression following wounding. The functional consequence of these observations was confirmed by demonstrating that 1,25D3 enabled keratinocytes to recognize microbial components through TLR2 and respond by cathelicidin production. Thus, we demonstrate what we believe to be a previously unexpected role for vitamin D3 in innate immunity, enabling keratinocytes to recognize and respond to microbes and to protect wounds against infection.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Epidermis/inmunología , Receptor Toll-Like 2/genética , Vitamina D/fisiología , Cicatrización de Heridas/inmunología , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/antagonistas & inhibidores , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/genética , Animales , Péptidos Catiónicos Antimicrobianos/genética , Calcitriol/farmacología , Células Epidérmicas , Epidermis/química , Expresión Génica/efectos de los fármacos , Humanos , Inmunidad Innata/genética , Queratinocitos/inmunología , Receptores de Lipopolisacáridos/genética , Ratones , Ratones Mutantes , Receptores de Calcitriol/antagonistas & inhibidores , Receptor Toll-Like 2/análisis , Receptor Toll-Like 2/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/genética , Catelicidinas
3.
J Bone Miner Res ; 22(9): 1329-37, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17539737

RESUMEN

UNLABELLED: We showed that the IGF-IR-null mutation in mature osteoblasts leads to less bone and decreased periosteal bone formation and impaired the stimulatory effects of PTH on osteoprogenitor cell proliferation and differentiation. INTRODUCTION: This study was carried out to examine the role of IGF-I signaling in mediating the actions of PTH on bone. MATERIALS AND METHODS: Three-month-old mice with an osteoblast-specific IGF-I receptor null mutation (IGF-IR OBKO) and their normal littermates were treated with vehicle or PTH (80 microg/kg body weight/d for 2 wk). Structural measurements of the proximal and midshaft of the tibia were made by microCT. Trabecular and cortical bone formation was measured by bone histomorphometry. Bone marrow stromal cells (BMSCs) were obtained to assess the effects of PTH on osteoprogenitor number and differentiation. RESULTS: The fat-free weight of bone normalized to body weight (FFW/BW), bone volume (BV/TV), and cortical thickness (C.Th) in both proximal tibia and shaft were all less in the IGF-IR OBKO mice compared with controls. PTH decreased FFW/BW of the proximal tibia more substantially in controls than in IGF-IR OBKO mice. The increase in C.Th after PTH in the proximal tibia was comparable in both control and IGF-IR OBKO mice. Although trabecular and periosteal bone formation was markedly lower in the IGF-IR OBKO mice than in the control mice, endosteal bone formation was comparable in control and IGF-IR OBKO mice. PTH stimulated endosteal bone formation only in the control animals. Compared with BMSCs from control mice, BMSCs from IGF-IR OBKO mice showed equal alkaline phosphatase (ALP)(+) colonies on day 14, but fewer mineralized nodules on day 28. Administration of PTH increased the number of ALP(+) colonies and mineralized nodules on days 14 and 28 in BMSCs from control mice, but not in BMSCs from IGF-IR OBKO mice. CONCLUSIONS: Our results indicate that the IGF-IR null mutation in mature osteoblasts leads to less bone and decreased bone formation, in part because of the requirement for the IGF-IR in mature osteoblasts to enable PTH to stimulate osteoprogenitor cell proliferation and differentiation.


Asunto(s)
Huesos/fisiología , Hormona Paratiroidea/fisiología , Receptor IGF Tipo 1/fisiología , Animales , Secuencia de Bases , Biomarcadores/metabolismo , Peso Corporal , Proliferación Celular , Células Cultivadas , Cartilla de ADN , Ratones , Ratones Noqueados , Mutación , Tamaño de los Órganos , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Transducción de Señal
4.
J Bone Miner Res ; 21(9): 1350-8, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16939393

RESUMEN

UNLABELLED: We showed that IGF-I deficiency impaired osteoclastogenesis directly and/or indirectly by altering the interaction between stromal/osteoblastic cells and osteoclast precursors, reducing RANKL and M-CSF production. These changes lead to impaired bone resorption, resulting in high BV/TV in IGF-I null mice. INTRODUCTION: Although IGF-I has been clearly identified as an important growth factor in regulating osteoblast function, information regarding its role in osteoclastogenesis is limited. Our study was designed to analyze the role of IGF-I in modulating osteoclastogenesis using IGF-I knockout mice (IGF-I(-/-)). MATERIALS AND METHODS: Trabecular bone volume (BV/TV), osteoclast number, and morphology of IGF-I(-/-) or wildtype mice (IGF-I(+/+)) were evaluated in vivo by histological analysis. Osteoclast precursors from these mice were cultured in the presence of RANKL and macrophage-colony stimulating factor (M-CSF) or co-cultured with stromal/osteoblastic cells from either genotype. Osteoclast formation was assessed by measuring the number of multinucleated TRACP+ cells and pit formation. The mRNA levels of osteoclast regulation markers were determined by quantitative RT-PCR. RESULTS: In vivo, IGF-I(-/-) mice have higher BV/TV and fewer (76% of IGF-I(+/+)) and smaller osteoclasts with fewer nuclei. In vitro, in the presence of RANKL and M-CSF, osteoclast number (55% of IGF-I(+/+)) and resorptive area (30% of IGF-I(+/+)) in osteoclast precursor cultures from IGF-I(-/-) mice were significantly fewer and smaller than that from the IGF-I(+/+) mice. IGF-I (10 ng/ml) increased the size, number (2.6-fold), and function (resorptive area, 2.7-fold) of osteoclasts in cultures from IGF-I(+/+) mice, with weaker stimulation in cultures from IGF-I(-/-) mice. In co-cultures of IGF-I(-/-) osteoblasts with IGF-I(+/+) osteoclast precursors, or IGF-I(+/+) osteoblasts with IGF-I(-/-) osteoclast precursors, the number of osteoclasts formed was only 11% and 48%, respectively, of that from co-cultures of IGF-I(+/+) osteoblasts and IGF-I(+/+) osteoclast precursors. In the long bones from IGF-I(-/-) mice, mRNA levels of RANKL, RANK, M-CSF, and c-fms were 55%, 33%, 60%, and 35% of that from IGF-I(+/+) mice, respectively. CONCLUSIONS: Our results indicate that IGF-I regulates osteoclastogenesis by promoting their differentiation. IGF-I is required for maintaining the normal interaction between the osteoblast and osteoclast to support osteoclastogenesis through its regulation of RANKL and RANK expression.


Asunto(s)
Células Madre Hematopoyéticas/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/farmacología , Factor I del Crecimiento Similar a la Insulina/fisiología , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Animales , Resorción Ósea/genética , Huesos/fisiología , Proteínas Portadoras/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Glicoproteínas/metabolismo , Células Madre Hematopoyéticas/metabolismo , Técnicas In Vitro , Factor I del Crecimiento Similar a la Insulina/genética , Factor Estimulante de Colonias de Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Factores de Transcripción NFATC/metabolismo , Osteoclastos/metabolismo , Osteoclastos/fisiología , Osteogénesis/genética , Osteoprotegerina , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Receptores de Calcitonina/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/fisiología
5.
Endocrinology ; 147(10): 4753-61, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16857753

RESUMEN

Although IGF-I has been identified as an important growth factor for the skeleton, the role of IGF-I on embryonic bone development remains unknown. Here we show that, in IGF-I-deficient (IGF-I(-/-)) mice, skeletal malformations, including short-limbed dwarfism, were evident at days post coitus (dpc) 14.5 to 18.5, accompanied by delays of mineralization in the spinal column, sternum, and fore paws. Reduced chondrocyte proliferation and increased chondrocyte apoptosis were identified in both the spinal ossification center and the growth plate of long bones. Abnormal chondrocyte differentiation and delayed initiation of mineralization was characterized by small size and fewer numbers of type X collagen expressing hypertrophic chondrocytes and lower osteocalcin expression. The Indian hedgehog-PTHrP feedback loop was altered; expression of Indian hedgehog was reduced in IGF-I(-/-) mice in long bones and in the spine, whereas expression of PTHrP was increased. Our results indicate that IGF-I plays an important role in skeletal development by promoting chondrocyte proliferation and maturation while inhibiting apoptosis to form bones of appropriate size and strength.


Asunto(s)
Desarrollo Óseo/fisiología , Huesos/embriología , Factor I del Crecimiento Similar a la Insulina/fisiología , Animales , Huesos/citología , Calcificación Fisiológica/genética , Calcificación Fisiológica/fisiología , Diferenciación Celular/fisiología , Proliferación Celular , Condrocitos/fisiología , Condrocitos/ultraestructura , Colágeno Tipo II/metabolismo , Factor I del Crecimiento Similar a la Insulina/deficiencia , Ratones , Ratones Noqueados , Microscopía Electrónica , Proteína Relacionada con la Hormona Paratiroidea/biosíntesis , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
J Endocrinol ; 189(2): 279-87, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16648295

RESUMEN

Parathyroid hormone (PTH) exerts both catabolic and anabolic actions on bone. Studies on the skeletal effects of PTH have seldom considered the effects of gender. Our study was designed to determine whether the response of mouse bone to PTH differed according to sex. As a first step, we analyzed gender differences with respect to bone mass and structural properties of 4 month old PTH treated (80 microg/kg per day for 2 weeks) male and female CD-1 mice. PTH significantly increased fat free weight/body weight, periosteal bone formation rate, mineral apposition rate, and endosteal single labeling surface, while significantly decreasing medullary area in male mice compared with vehicle treated controls, but induced no significant changes in female mice. We then analyzed the gender differences in bone marrow stromal cells (BMSC) isolated from 4 month old male and female CD-1 mice following treatment with PTH (80 microg/kg per day for 2 weeks). PTH significantly increased the osteogenic colony number and the alkaline phosphatase (ALP) activity (ALP/cell) by day 14 in cultures of BMSCs from male and female mice. PTH also increased the mRNA level of receptor activator of nuclear factor kappaB ligand in the bone tissue (marrow removed) of both females and males. However, PTH increased the mRNA levels of IGF-I and IGF-IR only in the bones of male mice. Our results indicate that on balance a 2-weeks course of PTH is anabolic on cortical bone in this mouse strain. These effects are more evident in the male mouse. These differences between male and female mice may reflect the greater response to PTH of IGF-I and IGF-IR gene expression in males enhancing the anabolic effect on cortical bone.


Asunto(s)
Huesos/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Hormona Paratiroidea/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Biomarcadores/análisis , Densidad Ósea/efectos de los fármacos , Densidad Ósea/fisiología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/enzimología , Células de la Médula Ósea/metabolismo , Huesos/anatomía & histología , Huesos/efectos de los fármacos , Proteínas Portadoras/análisis , Recuento de Células/métodos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Femenino , Factor I del Crecimiento Similar a la Insulina/análisis , Masculino , Glicoproteínas de Membrana/análisis , Ratones , Ratones Endogámicos , Tamaño de los Órganos/efectos de los fármacos , Tamaño de los Órganos/fisiología , Osteogénesis/efectos de los fármacos , Osteogénesis/fisiología , Hormona Paratiroidea/farmacología , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Receptor IGF Tipo 1/análisis , Factores Sexuales , Células del Estroma/efectos de los fármacos , Células del Estroma/enzimología , Células del Estroma/metabolismo , Tibia
7.
J Bone Miner Res ; 30(12): 2239-48, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26011431

RESUMEN

To investigate the role of IGF-I signaling in osterix (OSX)-expressing cells in the skeleton, we generated IGF-I receptor (IGF-IR) knockout mice ((OSX)IGF-IRKO) (floxed-IGF-IR mice × OSX promoter-driven GFP-labeled cre-recombinase [(OSX)GFPcre]), and monitored postnatal bone development. At day 2 after birth (P2), (OSX)GFP-cre was highly expressed in the osteoblasts in the bone surface of the metaphysis and in the prehypertrophic chondrocytes (PHCs) and inner layer of perichondral cells (IPCs). From P7, (OSX)GFP-cre was highly expressed in PHCs, IPCs, cartilage canals (CCs), and osteoblasts (OBs) in the epiphyseal secondary ossification center (SOC), but was only slightly expressed in the OBs in the metaphysis. Compared with the control mice, the IPC proliferation was decreased in the (OSX)IGF-IRKOs. In these mice, fewer IPCs invaded into the cartilage, resulting in delayed formation of the CC and SOC. Immunohistochemistry indicated a reduction of vessel number and lower expression of VEGF and ephrin B2 in the IPCs and SOC of (OSX)IGF-IRKOs. Quantitative real-time PCR revealed that the mRNA levels of the matrix degradation markers, MMP-9, 13 and 14, were decreased in the (OSX)IGF-IRKOs compared with the controls. The (OSX)IGF-IRKO also showed irregular morphology of the growth plate and less trabecular bone in the tibia and femur from P7 to 7 weeks, accompanied by decreased chondrocyte proliferation, altered chondrocyte differentiation, and decreased osteoblast differentiation. Our data indicate that during postnatal bone development, IGF-I signaling in OSX-expressing IPCs promotes IPC proliferation and cartilage matrix degradation and increases ephrin B2 production to stimulate vascular endothelial growth factor (VEGF) expression and vascularization. These processes are required for normal CC formation in the establishment of the SOC. Moreover, IGF-I signaling in the OSX-expressing PHC is required for growth plate maturation and osteoblast differentiation in the development of the metaphysis.


Asunto(s)
Desarrollo Óseo , Huesos/patología , Placa de Crecimiento/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Receptores de Somatomedina/metabolismo , Animales , Células de la Médula Ósea/citología , Huesos/metabolismo , Cartílago/metabolismo , Cartílago/patología , Diferenciación Celular , Proliferación Celular , Condrocitos/citología , Efrina-B2/metabolismo , Femenino , Fémur/patología , Eliminación de Gen , Genotipo , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica , Integrasas/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Noqueados , Osteoblastos/citología , Osteoblastos/metabolismo , Fenotipo , Regiones Promotoras Genéticas , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Somatomedina/genética , Transducción de Señal , Tibia/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
J Bone Miner Res ; 30(6): 1064-76, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25502173

RESUMEN

The primary goal of this study was to determine whether the IGF1R in mature osteoblasts and osteocytes was required for the catabolic actions of continuous parathyroid hormone (cPTH). Igf1r was deleted from male and female FVN/B mice by breeding with mice expressing cre recombinase under control of the osteocalcin promoter ((0CN) Igfr1(-/-) ). Littermates lacking the cre recombinase served as controls. PTH, 60 µg/kg/d, was administered continuously by Alzet minipumps for 4 weeks. Blood was obtained for indices of calcium metabolism. The femurs were examined by micro-computed tomography for structure, immunohistochemistry for IGF1R expression, histomorphometry for bone formation rates (BFR), mRNA levels by qPCR, and bone marrow stromal cell cultures (BMSC) for alkaline phosphatase activity (ALP(+) ), mineralization, and osteoblast-induced osteoclastogenesis. Whereas cPTH led to a reduction in trabecular bone volume/tissue volume (BV/TV) and cortical thickness in the control females, no change was found in the control males. Although trabecular BV/TV and cortical thickness were reduced in the (0CN) Igfr1(-/-) mice of both sexes, no further reduction after cPTH was found in the females, unlike the reduction in males. BFR was stimulated by cPTH in the controls but blocked by Igf1r deletion in the females. The (0CN) Igfr1(-/-) male mice showed a partial response. ALP(+) and mineralized colony formation were higher in BMSC from control males than from control females. These markers were increased by cPTH in both sexes, but BMSC from male (0CN) Igfr1(-/-) also were increased by cPTH, unlike those from female (0CN) Igfr1(-/-) . cPTH stimulated receptor activator of NF-κB ligand (RANKL) and decreased osteoprotegerin and alkaline phosphatase expression more in control female bone than in control male bone. Deletion of Igf1r blocked these effects of cPTH in the female but not in the male. However, PTH stimulation of osteoblast-driven osteoclastogenesis was blocked by deleting Igfr1 in both sexes. We conclude that cPTH is catabolic in female but not male mice. Moreover, IGF1 signaling plays a greater role in the skeletal actions of cPTH in the female mouse than in the male mouse, which may underlie the sex differences in the response to cPTH.


Asunto(s)
Desarrollo Óseo/efectos de los fármacos , Osteoblastos/metabolismo , Hormona Paratiroidea/farmacología , Receptor IGF Tipo 1/deficiencia , Caracteres Sexuales , Animales , Desarrollo Óseo/genética , Células Cultivadas , Femenino , Masculino , Ratones , Ratones Noqueados , Osteoblastos/efectos de la radiación , Hormona Paratiroidea/metabolismo
9.
J Bone Miner Res ; 19(3): 436-46, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15040832

RESUMEN

UNLABELLED: We showed that unloading markedly diminished the effects of IGF-I to activate its signaling pathways, and the disintegrin echistatin showed a similar block in osteoprogenitor cells. Furthermore, unloading decreased alphaVbeta3 integrin expression. These results show that skeletal unloading induces resistance to IGF-I by inhibiting activation of the IGF-I signaling pathways at least in part through downregulation of integrin signaling. INTRODUCTION: We have previously reported that skeletal unloading induces resistance to insulin-like growth factor-I (IGF-I) with respect to bone formation. However, the underlying mechanism remains unclear. The aim of this study was to clarify how skeletal unloading induces resistance to the effects of IGF-I administration in vivo and in vitro with respect to bone formation. MATERIALS AND METHODS: We first determined the response of bone to IGF-I administration in vivo during skeletal unloading. We then evaluated the response of osteoprogenitor cells isolated from unloaded bones to IGF-I treatment in vitro with respect to activation of the IGF-I signaling pathways. Finally we examined the potential role of integrins in mediating the responsiveness of osteoprogenitor cells to IGF-I. RESULTS: IGF-I administration in vivo significantly increased proliferation of osteoblasts. Unloading markedly decreased proliferation and blocked the ability of IGF-I to increase proliferation. On a cellular level, IGF-I treatment in vitro stimulated the activation of its receptor, Ras, ERK1/2 (p44/42 MAPK), and Akt in cultured osteoprogenitor cells from normally loaded bones, but these effects were markedly diminished in cells from unloaded bones. These results were not caused by altered phosphatase activity or changes in receptor binding to IGF-I. Inhibition of the Ras/MAPK pathway was more impacted by unloading than that of Akt. The disintegrin echistatin (an antagonist of the alphaVbeta3 integrin) blocked the ability of IGF-I to stimulate its receptor phosphorylation and osteoblast proliferation, similar to that seen in cells from unloaded bone. Furthermore, unloading significantly decreased the mRNA levels both of alphaV and beta3 integrin subunits in osteoprogenitor cells. CONCLUSION: These results indicate that skeletal unloading induces resistance to IGF-I by inhibiting the activation of IGF-I signaling pathways, at least in part, through downregulation of integrin signaling, resulting in decreased proliferation of osteoblasts and their precursors.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Osteogénesis , Animales , Secuencia de Bases , Células de la Médula Ósea/citología , Resorción Ósea , Regulación hacia Abajo , Suspensión Trasera/fisiología , Factor I del Crecimiento Similar a la Insulina/farmacología , Integrina alfaVbeta3/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Masculino , Datos de Secuencia Molecular , Osteoblastos/citología , Osteoclastos/citología , Osteogénesis/efectos de los fármacos , Péptidos/fisiología , ARN Mensajero/metabolismo , Ratas , Transducción de Señal
10.
J Bone Miner Res ; 29(8): 1900-13, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24677183

RESUMEN

Ephrin B2/EphB4 mediates interactions among osteoblasts (OBs), osteoclasts (OCLs), and chondrocytes to regulate their differentiation. We investigated the role of ephrin B2/EphB4 signaling in mediating the anabolic effects of insulin-like growth factor-I (IGF-I) and parathyroid hormone (PTH) on those cells and overall endochondral bone formation. Immunohistochemistry demonstrated that the expression of ephrin B2 in OBs, OCLs, and osteocytes, and the expression of EphB4 in OBs and osteocytes was dramatically decreased in global IGF-I knockout mice. Inactivation of EphB4 by EphB4 small, interfering RNA (siRNA) in cultured bone marrow stromal cells significantly decreased the mRNA levels of OB differentiation markers and abolished the stimulatory effects of IGF-I on these markers. Blocking the interaction of EphB4 and ephrin B2 in the OB-OCL cocultures with the EphB4 specific peptide TNYL-RAW or deletion of ephrin B2 in OCL prior to coculture led to fewer and smaller tartrate-resistant acid phosphatase (TRAP)-positive cells, decreased expression of OB differentiation markers, and blunted response to IGF-I for both OCL and OB differentiation. In the growth plate, both ephrin B2 and EphB4 are expressed in late stage proliferating and prehypertrophic chondrocytes, and their expression was decreased in mice lacking the IGF-I receptor specifically in chondrocytes. In vitro, blocking the interaction of EphB4 and ephrin B2 in chondrogenic ATDC5 cells with TNYL-RAW significantly decreased both basal and IGF1-induced expression of type II and type X collagen. In the cocultures of ATDC5 cells and spleen cells (osteoclast precursors), TNYL-RAW decreased the numbers of TRAP-positive cells and the expression of nuclear factor of activated T cells, cytoplasmic 1 (NFATc1) and receptor activator of NF-κB (RANK), and blocked their stimulation by IGF-I. Our data indicate that IGF-I/IGF-IR signaling promotes OB, OCL, and chondrocyte differentiation via ephrin B2/EphB4 mediated cell-cell communication.


Asunto(s)
Efrina-B2/genética , Factor I del Crecimiento Similar a la Insulina/fisiología , Receptor EphB4/genética , Transducción de Señal , Animales , Western Blotting , Comunicación Celular , Diferenciación Celular/genética , Efrina-B2/metabolismo , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Factor I del Crecimiento Similar a la Insulina/genética , Ratones , Ratones Noqueados , Osteoblastos/citología , Osteogénesis/genética , Receptor EphB4/metabolismo
11.
J Bone Miner Res ; 26(12): 2948-58, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21932337

RESUMEN

Integrin receptors bind extracellular matrix proteins, and this link between the cell membrane and the surrounding matrix may translate skeletal loading to biologic activity in osteoprogenitor cells. The interaction between integrin and growth factor receptors allows for mechanically induced regulation of growth factor signaling. Skeletal unloading leads to decreased bone formation and osteoblast proliferation that can be explained in part by a failure of insulin-like growth factor 1 (IGF-1) to activate its signaling pathways in unloaded bone. The aim of this study is to determine whether unloading-induced resistance is specific for IGF-1 or common to other skeletal growth factors, and to examine the regulatory role of integrins in IGF-1 signaling. Bone marrow osteoprogenitor (BMOp) cells were isolated from control or hindlimb suspended rats. Unloaded BMOp cells treated with IGF-1 failed to respond with increased proliferation, receptor phosphorylation, or signaling activation in the setting of intact ligand binding, whereas the platelet-derived growth factor (PDGF) response was fully intact. Pretreatment of control BMOp cells with an integrin inhibitor, echistatin, failed to disrupt PDGF signaling but blocked IGF-1 signaling. Recovery of IGF-1 signaling in unloaded BMOp cells followed the recovery of marked reduction in integrin expression induced by skeletal unloading. Selective targeting of integrin subunits with siRNA oligonucleotides revealed that integrin ß1 and ß3 are required for normal IGF-1 receptor phosphorylation. We conclude that integrins, in particular integrin ß3, are regulators of IGF-1, but not PDGF, signaling in osteoblasts, suggesting that PDGF could be considered for investigation in prevention and/or treatment of bone loss during immobilization and other forms of skeletal unloading.


Asunto(s)
Suspensión Trasera , Factor I del Crecimiento Similar a la Insulina/farmacología , Integrina beta1/metabolismo , Integrina beta3/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Integrina beta1/genética , Integrina beta3/genética , Péptidos y Proteínas de Señalización Intercelular , Ligandos , Masculino , Péptidos/farmacología , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor IGF Tipo 1/metabolismo
12.
J Bone Miner Res ; 26(7): 1437-46, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21312270

RESUMEN

Systemic derangements and perinatal death of generalized insulin-like growth factor 1 (IGF-1) and IGF-1 receptor (IGF-1R) knockout mice preclude definitive assessment of IGF-1R actions in growth-plate (GP) chondrocytes. We generated cartilage-specific Igf1r knockout ((Cart) Igf1r(-/-)) mice to investigate local control of chondrocyte differentiation in the GP by this receptor. These mice died shortly after birth and showed disorganized chondrocyte columns, delayed ossification and vascular invasion, decreased cell proliferation, increased apoptosis, and increased expression of parathyroid hormone-related protein (Pthrp) RNA and protein in their GPs. The increased Pthrp expression in the knockout GPs likely was due to an increase in gene transcription, as determined by the increased activity of a LacZ reporter that was inserted downstream of the endogenous PTHrP promoter and bred into the knockout mice. To circumvent the early death of (Cart) Igf1r(-/-) mice and investigate the role of IGF-1R during postnatal growth, we made tamoxifen (Tam)-inducible, cartilage-specific Igf1r knockout ((TamCart) Igf1r(-/-)) mice. At 2 weeks of age and 7 to 8 days after Tam injection, the (TamCart) Igf1r(-/-) mice showed growth retardation with a disorganized GP, reduced chondrocyte proliferation, decreased type 2 collagen and Indian Hedgehog (Ihh) expression, but increased expression of PTHrP. Consistent with in vivo observations, in vitro knockout of the Igf1r gene by adenoviral expression of Cre recombinase suppressed cell proliferation, promoted apoptosis, and increased Pthrp expression. Our data indicate that the IGF-1R in chondrocytes controls cell growth, survival, and differentiation in embryonic and postnatal GPs in part by suppression of Pthrp expression.


Asunto(s)
Condrocitos/metabolismo , Placa de Crecimiento/crecimiento & desarrollo , Proteínas Hedgehog/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Receptor IGF Tipo 1/metabolismo , Transducción de Señal , Animales , Animales Recién Nacidos , Apoptosis , Desarrollo Óseo , Huesos/embriología , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Condrocitos/citología , Eliminación de Gen , Regulación de la Expresión Génica , Placa de Crecimiento/citología , Placa de Crecimiento/metabolismo , Ratones , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA