Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Calcif Tissue Int ; 113(1): 96-109, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37243756

RESUMEN

Differentiation and optimal function of osteoblasts and osteoclasts are contingent on synthesis and maintenance of a healthy proteome. Impaired and/or altered secretory capacity of these skeletal cells is a primary driver of most skeletal diseases. The endoplasmic reticulum (ER) orchestrates the folding and maturation of membrane as well as secreted proteins at high rates within a calcium rich and oxidative organellar niche. Three ER membrane proteins monitor fidelity of protein processing in the ER and initiate an intricate signaling cascade known as the Unfolded Protein Response (UPR) to remediate accumulation of misfolded proteins in its lumen, a condition referred to as ER stress. The UPR aids in fine-tuning, expanding and/or modifying  the cellular proteome, especially in specialized secretory cells, to match everchanging physiologic cues and metabolic demands. Sustained activation of the UPR due to chronic ER stress, however, is known to hasten cell death and drive pathophysiology of several diseases. A growing body of evidence suggests that ER stress and an aberrant UPR may contribute to poor skeletal health and the development of osteoporosis. Small molecule therapeutics that target distinct components of the UPR may therefore have implications for developing novel treatment modalities relevant to the skeleton. This review summarizes the complexity of UPR actions in bone cells in the context of skeletal physiology and osteoporotic bone loss, and highlights the need for future mechanistic studies to develop novel UPR therapeutics that mitigate adverse skeletal outcomes.


Asunto(s)
Proteoma , Respuesta de Proteína Desplegada , Proteoma/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Transducción de Señal , Retículo Endoplásmico/metabolismo
2.
Curr Osteoporos Rep ; 16(2): 77-94, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29508144

RESUMEN

PURPOSE OF REVIEW: The international mouse phenotyping consortium (IMPC) is producing defined gene knockout mouse lines. Here, a phenotyping program is presented that is based on micro-computed tomography (µCT) assessment of distal femur and vertebra. Lines with significant variation undergo a computer-based bone histomorphometric analysis. RECENT FINDINGS: Of the 220 lines examined to date, approximately 15% have a significant variation (high or low) by µCT, most of which are not identified by the IMPC screen. Significant dimorphism between the sexes and bone compartments adds to the complexity of the skeletal findings. The µCT information that is posted at www.bonebase.org can group KOMP lines with similar morphological features. The histological data is presented in a graphic form that associates the cellular features with a specific anatomic group. The web portal presents a bone-centric view appropriate for the skeletal biologist/clinician to organize and understand the large number of genes that can influence skeletal health. Cataloging the relative severity of each variant is the first step towards compiling the dataset necessary to appreciate the full polygenic basis of degenerative bone disease.


Asunto(s)
Huesos/diagnóstico por imagen , Fémur/diagnóstico por imagen , Columna Vertebral/diagnóstico por imagen , Animales , Huesos/patología , Bases de Datos Factuales , Fémur/patología , Genotipo , Gestión de la Información , Ratones , Ratones Noqueados , Fenotipo , Desarrollo de Programa , Índice de Severidad de la Enfermedad , Caracteres Sexuales , Columna Vertebral/patología , Microtomografía por Rayos X
3.
J Cell Biochem ; 117(3): 599-611, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26252425

RESUMEN

The effect of targeted expression of an anabolic isoform of basic fibroblast growth factor (FGF2) in osteoblastic lineage on tibial fracture healing was assessed in mice. Closed fracture of the tibiae was performed in Col3.6-18 kDaFgf2-IRES-GFPsaph mice in which a 3.6 kb fragment of type I collagen promoter (Col3.6) drives the expression of only the 18 kD isoform of FGF2 (18 kDaFgf2/LMW) with green fluorescent protein-sapphire (GFPsaph) as well as Vector mice (Col3.6-IRES-GFPsaph, Vector) that did not harbor the FGF2 transgene. Radiographic, micro-CT, DEXA, and histologic analysis of fracture healing of tibiae harvested at 3, 10 and 20 days showed a smaller fracture callus but accelerated fracture healing in LMWTg compared with Vector mice. At post fracture day 3, FGF receptor 3 and Sox 9 mRNA were significantly increased in LMWTg compared with Vector. Accelerated fracture healing was associated with higher FGF receptor 1, platelet derived growth factors B, C, and D, type X collagen, vascular endothelial cell growth factor, matrix metalloproteinase 9, tartrate resistant acid phosphatase, cathepsin K, runt-related transcription factor-2, Osterix and Osteocalcin and lower Sox9, and type II collagen expression at 10 days post fracture. We postulate that overexpression of LMW FGF2 accelerated the fracture healing process due to its effects on factors that are important in chondrocyte and osteoblast differentiation and vascular invasion.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Curación de Fractura , Tibia/fisiopatología , Animales , Catepsina K/genética , Catepsina K/metabolismo , Colágeno Tipo II/metabolismo , Femenino , Factor 2 de Crecimiento de Fibroblastos/genética , Expresión Génica , Masculino , Ratones Transgénicos , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Factor de Transcripción SOX9/metabolismo , Fosfatasa Ácida Tartratorresistente/metabolismo , Tibia/diagnóstico por imagen , Tibia/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
Mamm Genome ; 27(7-8): 367-80, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27272104

RESUMEN

With aging, the skeleton experiences a number of changes, which include reductions in mass and changes in matrix composition, leading to fragility and ultimately an increase of fracture risk. A number of aspects of bone physiology are controlled by genetic factors, including peak bone mass, bone shape, and composition; however, forward genetic studies in humans have largely concentrated on clinically available measures such as bone mineral density (BMD). Forward genetic studies in rodents have also heavily focused on BMD; however, investigations of direct measures of bone strength, size, and shape have also been conducted. Overwhelmingly, these studies of the genetics of bone strength have identified loci that modulate strength via influencing bone size, and may not impact the matrix material properties of bone. Many of the rodent forward genetic studies lacked sufficient mapping resolution for candidate gene identification; however, newer studies using genetic mapping populations such as Advanced Intercrosses and the Collaborative Cross appear to have overcome this issue and show promise for future studies. The majority of the genetic mapping studies conducted to date have focused on younger animals and thus an understanding of the genetic control of age-related bone loss represents a key gap in knowledge.


Asunto(s)
Fracturas Óseas/genética , Osteoporosis/genética , Esqueleto/fisiopatología , Envejecimiento/genética , Envejecimiento/patología , Densidad Ósea , Mapeo Cromosómico , Fracturas Óseas/fisiopatología , Humanos , Osteoporosis/fisiopatología
5.
J Biol Chem ; 288(35): 25614-25625, 2013 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-23884415

RESUMEN

Notch receptors play a role in skeletal development and homeostasis, and Notch activation in undifferentiated and mature osteoblasts causes osteopenia. In contrast, Notch activation in osteocytes increases bone mass, but the mechanisms involved and exact functions of Notch are not known. In this study, Notch1 and -2 were inactivated preferentially in osteocytes by mating Notch1/2 conditional mice, where Notch alleles are flanked by loxP sequences, with transgenics expressing Cre directed by the Dmp1 (dentin matrix protein 1) promoter. Notch1/2 conditional null male and female mice exhibited an increase in trabecular bone volume due to an increase in osteoblasts and decrease in osteoclasts. In male null mice, this was followed by an increase in osteoclast number and normalization of bone volume. To activate Notch preferentially in osteocytes, Dmp1-Cre transgenics were crossed with Rosa(Notch) mice, where a loxP-flanked STOP cassette is placed between the Rosa26 promoter and Notch1 intracellular domain sequences. Dmp1-Cre(+/-);Rosa(Notch) mice exhibited an increase in trabecular bone volume due to decreased bone resorption and an increase in cortical bone due to increased bone formation. Biomechanical and chemical properties were not affected. Osteoprotegerin mRNA was increased, sclerostin and dickkopf1 mRNA were decreased, and Wnt signaling was enhanced in Dmp1-Cre(+/-);Rosa(Notch) femurs. Botulinum toxin A-induced muscle paralysis caused pronounced osteopenia in control mice, but bone mass was preserved in mice harboring the Notch activation in osteocytes. In conclusion, Notch plays a unique role in osteocytes, up-regulates osteoprotegerin and Wnt signaling, and differentially regulates trabecular and cortical bone homeostasis.


Asunto(s)
Remodelación Ósea , Osteocitos/metabolismo , Receptor Notch1/metabolismo , Receptor Notch2/metabolismo , Vía de Señalización Wnt , Animales , Enfermedades Óseas Metabólicas/inducido químicamente , Enfermedades Óseas Metabólicas/genética , Enfermedades Óseas Metabólicas/metabolismo , Enfermedades Óseas Metabólicas/patología , Toxinas Botulínicas Tipo A/efectos adversos , Toxinas Botulínicas Tipo A/farmacología , Femenino , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Ratones , Ratones Transgénicos , Fármacos Neuromusculares/efectos adversos , Fármacos Neuromusculares/farmacología , Osteocitos/patología , Osteoprotegerina/biosíntesis , Osteoprotegerina/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor Notch1/genética , Receptor Notch2/genética
6.
Stem Cells ; 30(2): 187-96, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22083974

RESUMEN

Adult mesenchymal progenitor cells have enormous potential for use in regenerative medicine. However, the true identity of the progenitors in vivo and their progeny has not been precisely defined. We hypothesize that cells expressing a smooth muscle α-actin promoter (αSMA)-directed Cre transgene represent mesenchymal progenitors of adult bone tissue. By combining complementary colors in combination with transgenes activating at mature stages of the lineage, we characterized the phenotype and confirmed the ability of isolated αSMA(+) cells to progress from a progenitor to fully mature state. In vivo lineage tracing experiments using a new bone formation model confirmed the osteogenic phenotype of αSMA(+) cells. In vitro analysis of the in vivo-labeled SMA9(+) cells supported their differentiation potential into mesenchymal lineages. Using a fracture-healing model, αSMA9(+) cells served as a pool of fibrocartilage and skeletal progenitors. Confirmation of the transition of αSMA9(+) progenitor cells to mature osteoblasts during fracture healing was assessed by activation of bone-specific Col2.3emd transgene. Our findings provide a novel in vivo identification of defined population of mesenchymal progenitor cells with active role in bone remodeling and regeneration.


Asunto(s)
Linaje de la Célula , Células Madre Mesenquimatosas/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Antígenos de Diferenciación/metabolismo , Células de la Médula Ósea/metabolismo , Regeneración Ósea , Remodelación Ósea , Diferenciación Celular , Femenino , Curación de Fractura , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Masculino , Ratones , Ratones Transgénicos , Fenotipo , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Tibia/patología
7.
Mol Ther ; 19(5): 960-8, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21343916

RESUMEN

Ex-vivo regional gene therapy with bone marrow cells (BMCs) overexpressing bone morphogenetic protein-2 (BMP-2) has demonstrated efficacy in healing critical sized bone defects in preclinical studies. The purpose of this preclinical study was to compare the osteoinductive potential of a novel "same day" ex-vivo regional gene therapy versus a traditional two-step approach, which involves culture expansion of the donor cells before implantation. In the "same day" strategy buffy coat cells were harvested from the rat bone marrow, transduced with a lentiviral vector-expressing BMP-2 for 1 hour and implanted into a rat femoral defect in the same sitting. There was no significant difference (P = 0.22) with respect to the radiographic healing rates between the femoral defects treated with the "same day" strategy (13/13; 100%) versus the traditional two-step approach (11/14; 78%). However, the femoral defects treated with the "same day" strategy induced earlier radiographic bone healing (P = 0.004) and higher bone volume (BV) [micro-computed tomography (micro-CT); P < 0.001]. The "same day" strategy represents a significant advance in the field of ex-vivo regional gene therapy because it offers a solution to limitations associated with the culture expansion process required in the traditional ex vivo approach. This strategy should be cost-effective when adapted for human use.


Asunto(s)
Células de la Médula Ósea/citología , Proteína Morfogenética Ósea 2/metabolismo , Regeneración Ósea/genética , Curación de Fractura/genética , Fracturas Óseas/terapia , Terapia Genética/métodos , Animales , Desarrollo Óseo , Células de la Médula Ósea/metabolismo , Proteína Morfogenética Ósea 2/genética , Trasplante Óseo/métodos , Células Cultivadas/virología , Ensayo de Inmunoadsorción Enzimática , Fracturas Óseas/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Vectores Genéticos/uso terapéutico , Lentivirus/genética , Ratas , Ratas Endogámicas Lew , Transducción Genética
8.
J Hand Surg Am ; 36(8): 1294-302, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21715102

RESUMEN

PURPOSE: New repair techniques for fragility fractures such as those of the distal radius require biomechanical justification. This study was conducted to investigate a technique using an expanding polymer bone cement to provide strength to a fracture repair. METHODS: Distal and proximal ends were isolated from 6 pairs of human radii (mean age 65). Transverse osteotomies were made near the head of each specimen. Paired specimens were repaired using 2 materials of differing polymer chemistries: polyurethane versus polymethylmethacrylate. Repaired specimens were subjected to failure tests in a cantilever beam configuration (distal, n = 6 per treatment) or pure tension (proximal, n = 5 per treatment). Cement penetration tests were conducted using a uniform open-cell model of cancellous bone. Baseline mechanical properties of the polyurethane cement were determined according to ASTM standards. RESULTS: Distal radii repaired with polyurethane bone cement withstood average shear stress 2.9 times as high as polymethylmethacrylate (0.91 vs 0.31 MPa). Peak tensile bending stress was 2.5 times as high in polyurethane (2.57 vs 1.02 MPa). Under pure tension, polyurethane-repaired samples failed at 0.83 MPa versus 0.74 MPa for polymethylmethacrylate. The polyurethane cement expanded to penetrate 49% farther into the trabeculae. The polyurethane cement had mean compressive yield stress of 20.3 MPa, compressive modulus of 754 MPa, ultimate tensile stress of 18.5 MPa, and tensile elastic modulus of 723 MPa. CONCLUSIONS: The biomechanical strength data indicate the potential of an expanding bone cement as a candidate strategy for fracture repair. Further evaluation might provide evidence for such an alternative repair strategy for fragility fractures, including those of the distal radius.


Asunto(s)
Cementos para Huesos/química , Fijación Interna de Fracturas/métodos , Poliuretanos/química , Fracturas del Radio/cirugía , Adulto , Anciano , Anciano de 80 o más Años , Fenómenos Biomecánicos , Cadáver , Femenino , Humanos , Masculino , Persona de Mediana Edad , Polimetil Metacrilato/química , Estrés Mecánico
9.
Methods Mol Biol ; 2230: 91-103, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33197010

RESUMEN

Given the prevalence and the scope of the personal and societal burden of osteoarthritis (OA), investigators continue to be deeply interested in understanding the pathogenic basis of disease and developing novel disease modifying OA therapies. Because joint trauma/injury is considered a leading predisposing factor in the development of OA, and since posttraumatic OA is one of the most common forms of OA in general, large animal and rodent models of knee injury that accurately recapitulate the OA disease process have become increasingly widespread over the past decade. To enable study in the context of defined genetic backgrounds, investigative teams have developed standardized protocols for injuring the mouse knee that aim to induce a reproducible degenerative process both in terms of severity and temporal pacing of disease progression. The destabilization of the medial meniscus (DMM) is one of the most commonly employed surgical procedure in rodents that reproducibly models posttraumatic OA and allows for the study of disease progression from initiation to end-stage disease. The description provided here sets the stage for both inexperienced and established investigators to employ the DMM procedure, or other similar surgical destabilization methods, to initiate the development of posttraumatic OA in the mouse. Successful application of this method provides a preclinical platform to study the mechanisms driving the pathogenesis of posttraumatic OA and for testing therapeutic strategies to treat it.


Asunto(s)
Cartílago Articular/crecimiento & desarrollo , Traumatismos de la Rodilla/cirugía , Meniscos Tibiales/cirugía , Osteoartritis/cirugía , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Traumatismos de la Rodilla/fisiopatología , Articulación de la Rodilla/fisiopatología , Articulación de la Rodilla/cirugía , Meniscos Tibiales/fisiopatología , Ratones , Osteoartritis/fisiopatología
10.
Bone ; 144: 115688, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33065355

RESUMEN

The IMPC/KOMP program provides the opportunity to screen mice harboring well defined gene-inactivation mutations in a uniform genetic background. The program performs a global tissue phenotyping survey that includes skeletal x-rays and bone density measurements. Because of the relative insensitivity of the two screening tests for detecting variance in bone architecture, we initiated a secondary screen based on µCT and a cryohistolomorphological workflow that was performed on the femur and vertebral compartments on 220 randomly selected knockouts (KOs) and 36 control bone samples over a 2 1/2 year collection period provided by one of the production/phenotyping centers. The performance of the screening protocol was designed to balance throughput and cost versus sensitivity and informativeness such that the output would be of value to the skeletal biology community. Here we report the reliability of this screening protocol to establish criteria for control skeletal variance at the architectural, dynamic and cellular histomorphometric level. Unexpected properties of the control population include unusually high variance in BV/TV in male femurs and greater bone formation and bone turnover rates in the female femur and vertebral trabeculae bone compartments. However, the manner for maintaining bone formation differed between these two bone sites. The vertebral compartment relies on maintaining a greater number of bone forming surfaces while the femoral compartment utilized more matrix production per cell. The comparison of the architectural properties obtained by µCT and histomorphology revealed significant differences in values for BV/TV, Tb.Th and Tb.N which is attributable to sampling density of the two methods. However, as a screening tool, expressing the ratio of KO to the control line as obtained by either method was remarkably similar. It identified KOs with significant variance which led to a more detailed histological analysis. Our findings are exemplified by the Efna4 KO, in which a high BV/TV was identified by µCT and confirmed by histomorphometry in the femur but not in the vertebral compartment. Dynamic labeling showed a marked increase in BFR which was attributable to increased labeling surfaces. Cellular analysis confirmed partitioning of osteoblast to labeling surfaces and a marked decrease in osteoclastic activity on both labeling and quiescent surfaces. This pattern of increased bone modeling would not be expected based on prior studies of the Ephrin-Ephrin receptor signaling pathways between osteoblasts and osteoclasts. Overall, our findings underscore why unbiased screening is needed because it can reveal unknown or unanticipated genes that impact skeletal variation.


Asunto(s)
Densidad Ósea , Fémur , Animales , Huesos/diagnóstico por imagen , Computadores , Femenino , Fémur/diagnóstico por imagen , Masculino , Ratones , Reproducibilidad de los Resultados
11.
J Nutr ; 140(8): 1495-501, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20534876

RESUMEN

A hallmark of energy restriction (ER) is a decrease in total body fat, which is thought to increase lifespan and maintain immune function. However, we have shown that during primary influenza infection, ER induces rapid weight loss, impairs natural killer (NK) cell function, and increases mortality in young and aged mice. To determine whether influenza-induced NK cell function could be restored in ER mice, young adult (6 mo) male C57BL/6 mice were fed an ER diet or re-fed (RF) control diet ad libitum for 2 wk before infection with PR8 influenza A. An initial hyperphagic response was observed in RF mice, characterized by increased food intake, rapid weight gain, and restoration of body fat and fat depots by 5-7 d of re-feeding to levels comparable to control ad libitum (AL) mice. Re-feeding improved survival and attenuated the decline in NK cell function during infection, evidenced by increased numbers, percentages, and CD69 expression by d 3 postinfection in RF mice. Interestingly, an altered metabolic phenotype was observed during infection of RF mice, with plasma leptin concentrations greater than in ER mice but less than in AL mice. In contrast, adiponectin concentrations of RF mice were lower than those of both ER and AL mice. These data suggest that re-feeding for a defined period before, and perhaps throughout, influenza season may provide the energy needed to counter the deleterious effects of ER on NK cell function, especially during exposure to newly emerging strains of influenza, for which vaccines are limited or unavailable.


Asunto(s)
Adiposidad , Restricción Calórica/efectos adversos , Alimentos , Células Asesinas Naturales/inmunología , Infecciones por Orthomyxoviridae/inmunología , Adipocitos , Adiponectina/sangre , Animales , Peso Corporal , Células de la Médula Ósea , Ingestión de Alimentos , Virus de la Influenza A , Leptina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/sangre , Infecciones por Orthomyxoviridae/patología
12.
J Bone Miner Res ; 35(1): 130-142, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31487060

RESUMEN

CD55 is a glycosylphosphatidylinositol (GPI)-anchored protein that regulates complement-mediated and innate and adaptive immune responses. Although CD55 is expressed in various cell types in the bone marrow, its role in bone has not been investigated. In the current study, trabecular bone volume measured by µCT in the femurs of CD55KO female mice was increased compared to wild type (WT). Paradoxically, osteoclast number was increased in CD55KO with no differences in osteoblast parameters. Osteoclasts from CD55KO mice exhibited abnormal actin-ring formation and reduced bone-resorbing activity. Moreover, macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL) treatment failed to activate Rac guanosine triphosphatase (GTPase) in CD55KO bone marrow macrophage (BMM) cells. In addition, apoptotic caspases activity was enhanced in CD55KO, which led to the poor survival of mature osteoclasts. Our results imply that CD55KO mice have increased bone mass due to defective osteoclast resorbing activity resulting from reduced Rac activity in osteoclasts. We conclude that CD55 plays an important role in the survival and bone-resorption activity of osteoclasts through regulation of Rac activity. © 2019 American Society for Bone and Mineral Research.


Asunto(s)
Resorción Ósea , Osteoclastos , Animales , Células de la Médula Ósea , Diferenciación Celular , Femenino , Factor Estimulante de Colonias de Macrófagos , Ratones , Osteoblastos , Ligando RANK , Transducción de Señal
13.
Ann N Y Acad Sci ; 1460(1): 57-67, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31596513

RESUMEN

Following anterior cruciate ligament (ACL) reconstruction surgery, a staged repair response occurs where cells from outside the tendon graft participate in tunnel integration. The mechanisms that regulate this process, including the specific cellular origin, are poorly understood. Embryonic cells expressing growth and differentiation factor 5 (GDF5) give rise to several mesenchymal tissues in the joint and epiphyses. We hypothesized that cells from a GDF5 origin, even in the adult tissue, would give rise to cells that contribute to the stages of repair. ACLs were reconstructed in Gdf5-Cre;R26R-tdTomato lineage tracing mice to monitor the contribution of Gdf5-Cre;tdTom+ cells to the tunnel integration process. Anterior-posterior drawer tests demonstrated 58% restoration in anterior-posterior stability. Gdf5-Cre;tdTom+ cells within the epiphyseal bone marrow adjacent to tunnels expanded in response to the injury by 135-fold compared with intact controls to initiate tendon-to-bone attachments. They continued to mature the attachments yielding zonal insertion sites at 4 weeks with collagen fibers spanning across unmineralized and mineralized fibrocartilage and anchored to the adjacent bone. The zonal attachments possessed tidemarks with concentrated alkaline phosphatase activity similar to native entheses. This study established that mesenchymal cells from a GDF5 origin can contribute to zonal tendon-to-bone attachments within bone tunnels following ACL reconstruction.


Asunto(s)
Reconstrucción del Ligamento Cruzado Anterior , Huesos/patología , Factor 5 de Diferenciación de Crecimiento/metabolismo , Tendones/patología , Animales , Médula Ósea/patología , Muerte Celular , Epífisis/patología , Integrasas/metabolismo , Ratones Transgénicos
14.
J Orthop Res ; 38(11): 2350-2361, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32141629

RESUMEN

Fracture healing involves interactions of different cell types, driven by various growth factors, and signaling cascades. Periosteal mesenchymal progenitor cells give rise to the majority of osteoblasts and chondrocytes in a fracture callus. Notch signaling has emerged as an important regulator of skeletal cell proliferation and differentiation. We investigated the effects of Notch signaling during the fracture healing process. Increased Notch signaling in osteochondroprogenitor cells driven by overexpression of Notch1 intracellular domain (NICD1) (αSMACreERT2 mice crossed with Rosa-NICD1) during fracture resulted in less cartilage, more mineralized callus tissue, and stronger and stiffer bones after 3 weeks. Periosteal cells overexpressing NICD1 showed increased proliferation and migration in vitro. In vivo data confirmed that increased Notch1 signaling caused expansion of alpha-smooth muscle actin (αSMA)-positive cells and their progeny including αSMA-derived osteoblasts in the callus without affecting osteoclast numbers. In contrast, anti-NRR1 antibody treatment to inhibit Notch1 signaling resulted in increased callus cartilage area, reduced callus bone mass, and reduced biomechanical strength. Our study shows a positive effect of induced Notch1 signaling on the fracture healing process, suggesting that stimulating the Notch pathway could be beneficial for fracture repair.


Asunto(s)
Curación de Fractura , Receptor Notch1/metabolismo , Animales , Femenino , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Receptor Notch1/antagonistas & inhibidores
15.
J Cell Biochem ; 108(3): 621-30, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19670271

RESUMEN

Osteocytes are the most abundant osteoblast lineage cells within the bone matrix. They respond to mechanical stimulation and can participate in the release of regulatory proteins that can modulate the activity of other bone cells. We hypothesize that neuropeptide Y (NPY), a neurotransmitter with regulatory functions in bone formation, is produced by osteocytes and can affect osteoblast activity. To study the expression of NPY by the osteoblast lineage cells, we utilized transgenic mouse models in which we can identify and isolate populations of osteoblasts and osteocytes. The Col2.3GFP transgene is active in osteoblasts and osteocytes, while the DMP1 promoter drives green fluorescent protein (GFP) expression in osteocytes. Real-time PCR analysis of RNA from the isolated populations of cells derived from neonatal calvaria showed higher NPY mRNA in the preosteocytes/osteocytes fraction compared to osteoblasts. NPY immunostaining confirmed the strong expression of NPY in osteocytes (DMP1GFP(+)), and lower levels in osteoblasts. In addition, the presence of NPY receptor Y1 mRNA was detected in cavaria and long bone, as well as in primary calvarial osteoblast cultures, whereas Y2 mRNA was restricted to the brain. Furthermore, NPY expression was reduced by 30-40% in primary calvarial cultures when subjected to fluid shear stress. In addition, treatment of mouse calvarial osteoblasts with exogenous NPY showed a reduction in the levels of intracellular cAMP and markers of osteoblast differentiation (osteocalcin, BSP, and DMP1). These results highlight the potential regulation of osteoblast lineage differentiation by local NPY signaling.


Asunto(s)
Neuropéptido Y/metabolismo , Osteocitos/metabolismo , Animales , Linaje de la Célula/efectos de los fármacos , Células Cultivadas , AMP Cíclico/metabolismo , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica/efectos de los fármacos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Ratones , Ratones Transgénicos , Neuropéptido Y/genética , Neuropéptido Y/farmacología , Osteocitos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Neuropéptido Y/genética , Receptores de Neuropéptido Y/metabolismo , Resistencia al Corte , Cráneo/citología , Cráneo/efectos de los fármacos , Cráneo/metabolismo
16.
Nat Genet ; 51(2): 258-266, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30598549

RESUMEN

Osteoporosis is a common aging-related disease diagnosed primarily using bone mineral density (BMD). We assessed genetic determinants of BMD as estimated by heel quantitative ultrasound in 426,824 individuals, identifying 518 genome-wide significant loci (301 novel), explaining 20% of its variance. We identified 13 bone fracture loci, all associated with estimated BMD (eBMD), in ~1.2 million individuals. We then identified target genes enriched for genes known to influence bone density and strength (maximum odds ratio (OR) = 58, P = 1 × 10-75) from cell-specific features, including chromatin conformation and accessible chromatin sites. We next performed rapid-throughput skeletal phenotyping of 126 knockout mice with disruptions in predicted target genes and found an increased abnormal skeletal phenotype frequency compared to 526 unselected lines (P < 0.0001). In-depth analysis of one gene, DAAM2, showed a disproportionate decrease in bone strength relative to mineralization. This genetic atlas provides evidence linking associated SNPs to causal genes, offers new insight into osteoporosis pathophysiology, and highlights opportunities for drug development.


Asunto(s)
Densidad Ósea/genética , Predisposición Genética a la Enfermedad/genética , Osteoporosis/genética , Adulto , Anciano , Animales , Femenino , Fracturas Óseas/genética , Estudio de Asociación del Genoma Completo/métodos , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Fenotipo , Polimorfismo de Nucleótido Simple/genética
18.
Endocrinology ; 149(8): 4009-15, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18467443

RESUMEN

PTH is the only currently available anabolic therapy for osteoporosis. In clinical practice, the skeletal response to PTH varies and because therapy is limited to 2 yr, approaches to maximize the therapeutic response are desirable. Rac2 is a small GTPase that is expressed only in hematopoietic tissue. Rac2(-/-) mice have a slight increase in bone mass and osteoclasts isolated from these animals have reduced basal resorptive activity and reduced chemotaxis. To evaluate the anabolic response to PTH in Rac2(-/-) mice, we treated 18 Rac2(-/-) and 17 control, age-matched wild-type animals once daily for 28 d with 80 ng/g body weight of h(1-34)PTH. Treatment resulted in significantly greater increments in spinal, femur, and total bone density in the Rac2(-/-) as compared with wild-type animals. Microcomputed tomography analysis demonstrated greater increases in trabecular thickness and cortical thickness in the knockout mice. Interestingly, histomorphometric analysis showed an equivalent increase in osteoblast and osteoclast number in response to PTH treatment in both groups of animals. However, as judged by changes in serum markers, the resorptive response to PTH was impaired. Thus, telopeptide of type 1 collagen was 15.9+/-6.9 ng/ml after PTH treatment in the knockout animals and 26.8+/-11.1 ng/ml in the PTH-treated wild-type group. In contrast, serum aminoterminal propeptide of type 1 collagen and osteocalcin were equivalent in both groups. We conclude that, in the genetic absence of Rac2, the anabolic response to PTH is increased. This appears to be due to attenuated resorptive activity of osteoclasts.


Asunto(s)
Huesos/efectos de los fármacos , Huesos/metabolismo , Hormona Paratiroidea/farmacología , Proteínas de Unión al GTP rac/genética , Anabolizantes/farmacología , Animales , Densidad Ósea/efectos de los fármacos , Densidad Ósea/genética , Remodelación Ósea/efectos de los fármacos , Remodelación Ósea/genética , Recuento de Células , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteocalcina/sangre , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Fragmentos de Péptidos/sangre , Procolágeno/sangre , Regulación hacia Arriba/efectos de los fármacos , Proteína RCA2 de Unión a GTP
19.
Bone ; 43(2): 264-273, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18499553

RESUMEN

Matricellular proteins play a unique role in the skeleton as regulators of bone remodeling, and the matricellular protein osteonectin (SPARC, BM-40) is the most abundant non-collagenous protein in bone. In the absence of osteonectin, mice develop progressive low turnover osteopenia, particularly affecting trabecular bone. Polymorphisms in a regulatory region of the osteonectin gene are associated with bone mass in a subset of idiopathic osteoporosis patients, and these polymorphisms likely regulate osteonectin expression. Thus it is important to determine how osteonectin gene dosage affects skeletal function. Moreover, intermittent administration of parathyroid hormone (PTH) (1-34) is the only anabolic therapy approved for the treatment of osteoporosis, and it is critical to understand how modulators of bone remodeling, such as osteonectin, affect skeletal response to anabolic agents. In this study, 10 week old female wild type, osteonectin-haploinsufficient, and osteonectin-null mice (C57Bl/6 genetic background) were given 80 microg/kg body weight/day PTH(1-34) for 4 weeks. Osteonectin gene dosage had a profound effect on bone microarchitecture. The connectivity density of trabecular bone in osteonectin-haploinsufficient mice was substantially decreased compared with that of wild type mice, suggesting compromised mechanical properties. Whereas mice of each genotype had a similar osteoblastic response to PTH treatment, the osteoclastic response was accentuated in osteonectin-haploinsufficient and osteonectin-null mice. Eroded surface and osteoclast number were significantly higher in PTH-treated osteonectin-null mice, as was endosteal area. In vitro studies confirmed that PTH induced the formation of more osteoclast-like cells in marrow from osteonectin-null mice compared with wild type. PTH treated osteonectin-null bone marrow cells expressed more RANKL mRNA compared with wild type. However, the ratio of RANKL:OPG mRNA was somewhat lower in PTH treated osteonectin-null cultures. Increased expression of RANKL in response to PTH could contribute to the accentuated osteoclastic response in osteonectin-/- mice, but other mechanisms are also likely to be involved. The molecular mechanisms by which PTH elicits bone anabolic vs. bone catabolic effects remain poorly understood. Our results imply that osteonectin levels may play a role in modulating the balance of bone formation and resorption in response to PTH.


Asunto(s)
Huesos/efectos de los fármacos , Huesos/patología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteonectina/deficiencia , Hormona Paratiroidea/farmacología , Animales , Densidad Ósea/efectos de los fármacos , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Femenino , Fémur/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos/efectos de los fármacos , Osteoclastos/citología , Osteonectina/metabolismo , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Ligando RANK/genética , Ligando RANK/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Columna Vertebral/efectos de los fármacos , Tomografía Computarizada por Rayos X
20.
Bone ; 43(1): 101-109, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18460422

RESUMEN

ICER is a member of the CREM family of basic leucine zipper transcription factors that acts as a dominant negative regulator of gene transcription. Four different isoforms of ICER (I, Igamma, II and IIgamma) are transcribed from the P2 promoter of the Crem gene. We previously found that each of the ICER isoforms is induced by parathyroid hormone in osteoblasts. The goal of the present study was to assess the function of ICER in bone by overexpressing ICER in osteoblasts of transgenic mice. ICER I and ICER II cDNAs, each containing an N-terminal FLAG epitope tag, were cloned downstream of a fragment containing 3.6 kb of the rat Col1a1 promoter and most of the rat Col1a1 first intron to produce pOBCol3.6-ICER I and pOBCol3.6-ICER II transgenes, respectively. Multiple lines of mice were generated bearing the ICER I and ICER II transgenes. At 8 weeks of age, ICER I and ICER II transgenic mice had lower body weights and decreased bone mineral density of femurs and vertebrae. Further studies were done with ICER I transgenic mice, which had greatly reduced trabecular bone volume and a markedly decreased bone formation rate in femurs. Osteoblast differentiation and osteocalcin expression were reduced in ex vivo bone marrow cultures from ICER I transgenic mice. ICER I antagonized the activity of ATF4 at its consensus DNA binding site in the osteocalcin promoter in vitro. Thus, transgenic mice with osteoblast-targeted overexpression of ICER exhibited osteopenia caused primarily by reduced bone formation. We speculate that ICER regulates the activity and/or expression of ATF/CREB factors required for normal bone formation.


Asunto(s)
Enfermedades Óseas Metabólicas/genética , Modulador del Elemento de Respuesta al AMP Cíclico/genética , Osteoblastos/metabolismo , Células 3T3 , Animales , Densidad Ósea/genética , Diferenciación Celular , Línea Celular , Humanos , Ratones , Ratones Transgénicos , Osteoblastos/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA