Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Transl Med ; 11(3): e353, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33783995

RESUMEN

BACKGROUND: Clear cell renal cell carcinoma (ccRCC) has gender differences, with the androgen receptor (AR) linked positively with metastasis to the lung. Its linkage to ccRCC bone metastases (RBMs), however, remains unclear. METHODS: In the current study, five human RCC and five RCC bone metastasis tissues were deeply sequenced using Arraystar human circRNA V2.0 microarray. We conducted gain-of-function screening in vitro and in vivo to elucidate the AR's role in the RBM. Loss/gain-of-function was also implemented to verify the roles of related non-coding RNAs and proteins. RESULTS: We uncovered that RBM also has a gender difference showing higher AR expression may be linked to fewer RBMs, which might involve suppressing osteolytic formation. Mechanism dissection indicates that AR can decrease the circular RNA EXOC7 (circEXOC7), expression via enhancing transcription of DHX9, a regulatory protein in circRNA biogenesis. The circEXOC7 can sponge/suppress miR-149-3p resulting in suppressing the CSF1 expression by directly binding to the 3'UTR region of CSF1 mRNA. Results from clinical epidemiological surveys also found that AR has a positive correlation with miR-149-3p and a negative correlation with CSF1 in AR-positive ccRCC tissues. Preclinical studies with Balb/c nude mouse model also validated that targeting this newly verified AR/DHX9/circEXOC7/miR-149-3p/CSF1 signaling via altering circEXOC7 or AR could lead to suppressing the RBM progression. CONCLUSIONS: These data showed that AR/DHX9/circEXOC7/miR-149-3p/CSF1 signaling acts as a valuable feature in the bone metastasis of renal cancer, which may benefit in suppressing the RBM progression.


Asunto(s)
Neoplasias Óseas/secundario , Carcinoma de Células Renales/patología , Neoplasias Renales/patología , ARN Circular/genética , Receptores Androgénicos/genética , Proteínas de Transporte Vesicular/genética , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/prevención & control , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Osteólisis/genética , Osteólisis/metabolismo , ARN Circular/metabolismo , Receptores Androgénicos/metabolismo , Transducción de Señal , Proteínas de Transporte Vesicular/metabolismo
2.
Aging Cell ; 20(1): e13288, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33336885

RESUMEN

Morphological change is an explicit characteristic of cell senescence, but the underlying mechanisms remains to be addressed. Here, we demonstrated, after a survey of various actin-binding proteins, that the post-translational up-regulation of cofilin-1 was essential for the reduced rate of actin depolymerization morphological enlargement in senescent cells. Additionally, up-regulated cofilin-1 mainly existed in the serine-3 phosphorylated form, according to the 2D gel immunoblotting assay. The up-regulation of cofilin-1 was also detected in aged mammalian tissues. The over-expression of wild-type cofilin-1 and constitutively phosphorylated cofilin-1 promoted cell senescence with an increased cell size. Additionally, senescent phenotypes were also reduced by knockdown of total cofilin-1, which led to a decrease in phosphorylated cofilin-1. The senescence induced by the over-expression of cofilin-1 was dependent on p27Kip1 , but not on the p53 and p16INK4 expressions. The knockdown of p27Kip1 alleviated cell senescence induced by oxidative stress or replicative stress. We also found that the over-expression of cofilin-1 induced the expression of p27Kip1 through transcriptional suppression of the transcriptional enhancer factors domain 1 (TEAD1) transcription factor. The TEAD1 transcription factor played a transrepressive role in the p27Kip1  gene promoter, as determined by the promoter deletion reporter gene assay. Interestingly, the down-regulation of TEAD1 was accompanied by the up-regulation of cofilin-1 in senescence. The knockdown and restoration of TEAD1 in young cells and old cells could induce and inhibit p27Kip1 and senescent phenotypes, respectively. Taken together, the current data suggest that cofilin-1/TEAD1/p27Kip1 signaling is involved in senescence-related morphological change and growth arrest.


Asunto(s)
Cofilina 1/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Senescencia Celular , Humanos , Regulación hacia Arriba
3.
Bone ; 137: 115391, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32360587

RESUMEN

Bone fracture is accompanied by trauma, mechanical stresses, and inflammation - conditions known to induce the mitochondrial permeability transition. This phenomenon occurs due to opening of the mitochondrial permeability transition pore (MPTP) promoted by cyclophilin D (CypD). MPTP opening leads to more inflammation, cell death and potentially to disruption of fracture repair. Here we performed a proof-of-concept study and tested a hypothesis that protecting mitochondria from MPTP opening via inhibition of CypD improves fracture repair. First, our in vitro experiments indicated pro-osteogenic and anti-inflammatory effects in osteoprogenitors upon CypD knock-out or pharmacological inhibition. Using a bone fracture model in mice, we observed that bone formation and biomechanical properties of repaired bones were significantly increased in CypD knock-out mice or wild type mice treated with a CypD inhibitor, NIM811, when compared to controls. These effects were evident in young male but not female mice, however in older (13 month-old) female mice bone formation was also increased during fracture repair. In contrast to global CypD knock-out, mesenchymal lineage-specific (Prx1-Cre driven) CypD deletion did not result in improved fracture repair. Our findings implicate MPTP in bone fracture and suggest systemic CypD inhibition as a modality to promote fracture repair.


Asunto(s)
Fracturas Óseas , Necrosis por Permeabilidad de la Transmembrana Mitocondrial , Animales , Peptidil-Prolil Isomerasa F , Femenino , Masculino , Ratones , Ratones Noqueados , Proteínas de Transporte de Membrana Mitocondrial
5.
EMBO Rep ; 21(4): e48467, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32052578

RESUMEN

The androgen receptor (AR) has been linked to bladder cancer (BCa) progression, but if this involves circular RNAs (circRNAs) remains unclear. Here, we find that AR alters the levels of circRNA-FNTA (circFNTA) to increase BCa cell invasion and chemo-resistance. Mechanistically, AR represses the RNA editing gene ADAR2 via direct binding to its 5' promoter region to increase circFNTA levels, which then sponges the microRNA miR-370-3p to increase the expression of its host gene FNTA. This AR-mediated ADAR2/circFNTA/miR-370-3p/FNTA pathway then activates KRAS signaling to alter BCa cell invasion and chemo-sensitivity to cisplatin. A clinical BCa sample survey shows that circFNTA expression is elevated in BCa tissues, and results from a BCa mouse model indicate that depletion of circFNTA leads to the suppression of BCa metastases and increased cisplatin chemo-sensitivity. Together, based on our results using multiple BCa cell lines and an in vivo mouse model we suggest that targeting this newly identified AR/ADAR2/circFNTA/miR-370-3p/FNTA/KRAS axis may lead to the development of therapies to suppress BCa metastasis and to increase its chemo-sensitivity.


Asunto(s)
Transferasas Alquil y Aril/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , ARN Circular/genética , Receptores Androgénicos/metabolismo , Neoplasias de la Vejiga Urinaria , Animales , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Ratones , Proteínas Proto-Oncogénicas p21(ras)/genética , Receptores Androgénicos/genética , Neoplasias de la Vejiga Urinaria/genética
6.
Nat Mater ; 18(6): 627-637, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31114073

RESUMEN

Cells are transplanted to regenerate an organs' parenchyma, but how transplanted parenchymal cells induce stromal regeneration is elusive. Despite the common use of a decellularized matrix, little is known as to the pivotal signals that must be restored for tissue or organ regeneration. We report that Alx3, a developmentally important gene, orchestrated adult parenchymal and stromal regeneration by directly transactivating Wnt3a and vascular endothelial growth factor. In contrast to the modest parenchyma formed by native adult progenitors, Alx3-restored cells in decellularized scaffolds not only produced vascularized stroma that involved vascular endothelial growth factor signalling, but also parenchymal dentin via the Wnt/ß-catenin pathway. In an orthotopic large-animal model following parenchyma and stroma ablation, Wnt3a-recruited endogenous cells regenerated neurovascular stroma and differentiated into parenchymal odontoblast-like cells that extended the processes into newly formed dentin with a structure-mechanical equivalency to native dentin. Thus, the Alx3-Wnt3a axis enables postnatal progenitors with a modest innate regenerative capacity to regenerate adult tissues. Depleted signals in the decellularized matrix may be reinstated by a developmentally pivotal gene or corresponding protein.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Tejido Parenquimatoso/fisiología , Diente/citología , Diente/embriología , Adolescente , Animales , Femenino , Proteínas de Homeodominio/genética , Humanos , Incisivo/citología , Incisivo/embriología , Ratones Endogámicos , Tercer Molar/citología , Técnicas de Cultivo de Órganos , Tejido Parenquimatoso/citología , Embarazo , Regiones Promotoras Genéticas , Regeneración , Células del Estroma/fisiología , Porcinos , Factor A de Crecimiento Endotelial Vascular/genética , Proteína Wnt3A/genética , Proteína Wnt3A/metabolismo
7.
Diabetologia ; 62(5): 822-834, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30824970

RESUMEN

AIMS/HYPOTHESIS: We previously reported that exposure to antibodies neutralising serpin B13, a protease inhibitor expressed in exocrine pancreatic ducts, promotes beta cell proliferation, underscoring the importance of a functional relationship between exocrine and endocrine pancreas. The aim of the present study was to identify the molecular events that link inhibition of serpin B13 to islet cell proliferation. METHODS: We used an in vitro culture system consisting of isolated pancreatic islets, an extract of pancreatic ductal epithelium and a monoclonal antibody (mAb) to serpin B13 or IgG isotype control. In vivo studies involved treatment of mice with these mAbs. RESULTS: The catalytic activity of cathepsin L (CatL), a cysteine protease target of serpin B13, was augmented in the pancreas of mice injected with serpin B13 mAb. Furthermore, the addition of serpin B13 mAb to the islets, together with the pancreatic ductal epithelium lysate, caused CatL-dependent cleavage of E-cadherin and concomitant upregulation of REG genes, ultimately leading to beta cell proliferation. Direct blockade of E-cadherin with mAb also markedly enhanced REG gene induction, while chemical inhibition of ß-catenin, a binding target of E-cadherin, prevented the serpin B13 mAb-induced upregulation of REG genes. CONCLUSIONS/INTERPRETATION: Our work implicates the CatL-E-cadherin-REG pathway in the regulation of islet cell proliferation in response to signals generated in exocrine pancreatic tissue and demonstrates that protease activity may promote adaptive changes in the islets. DATA AVAILABILITY: Microarray data that support the findings of this study have been deposited in Gene Expression Omnibus (GEO) with the accession no. GSE125151.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Catepsina L/metabolismo , Islotes Pancreáticos/metabolismo , Serpinas/metabolismo , Animales , Anticuerpos Monoclonales , Proliferación Celular , Femenino , Expresión Génica , Células HEK293 , Humanos , Islotes Pancreáticos/citología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Páncreas Exocrino/metabolismo , Conductos Pancreáticos/citología , Transducción de Señal , alfa 1-Antitripsina/metabolismo
8.
Biomacromolecules ; 19(1): 71-84, 2018 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-29227674

RESUMEN

Drug delivery to bone is challenging, whereby drug distribution is commonly <1% of injected dose, despite development of several bone-targeted drug delivery systems specific to hydroxyapatite. These bone-targeted drug delivery systems still suffer from poor target cell localization within bone, as at any given time overall bone volume is far greater than acutely remodeling bone volume, which harbors relevant cell targets (osteoclasts or osteoblasts). Thus, there exists a need to target bone-acting drugs specifically to sites of bone remodeling. To address this need, this study synthesized oligo(ethylene glycol) copolymers based on a peptide with high affinity to tartrate-resistant acid phosphatase (TRAP), an enzyme deposited by osteoclasts during the bone resorption phase of bone remodeling, which provides greater specificity relevant for bone cell drugging. Gradient and random peptide orientations, as well as polymer molecular weights, were investigated. TRAP-targeted, high molecular weight (Mn) random copolymers exhibited superior accumulation in remodeling bone, where fracture accumulation was observed for at least 1 week and accounted for 14% of tissue distribution. Intermediate and low Mn random copolymer accumulation was lower, indicating residence time depends on Mn. High Mn gradient polymers were cleared, with only 2% persisting at fractures after 1 week, suggesting TRAP binding depends on peptide density. Peptide density and Mn are easily modified in this versatile targeting platform, which can be applied to a range of bone drug delivery applications.


Asunto(s)
Sistemas de Liberación de Medicamentos , Péptidos/metabolismo , Polímeros/farmacocinética , Acrilamida/química , Animales , Remodelación Ósea , Células Cultivadas , Femenino , Colorantes Fluorescentes/química , Humanos , Masculino , Ratones Endogámicos C57BL , Peso Molecular , Osteoclastos/enzimología , Péptidos/química , Polímeros/química , Fosfatasa Ácida Tartratorresistente/metabolismo , Distribución Tisular
9.
ACS Nano ; 11(9): 9445-9458, 2017 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-28881139

RESUMEN

Despite several decades of progress, bone-specific drug delivery is still a major challenge. Current bone-acting drugs require high-dose systemic administration which decreases therapeutic efficacy and increases off-target tissue effects. Here, a bone-targeted nanoparticle (NP) delivery system for a ß-catenin agonist, 3-amino-6-(4-((4-methylpiperazin-1-yl)sulfonyl)phenyl)-N-(pyridin-3-yl)pyrazine-2-carboxamide, a glycogen synthase kinase 3 beta (GSK-3ß) inhibitor, was developed to enhance fracture healing. The GSK-3ß inhibitor loading capacity was found to be 15 wt % within highly stable poly(styrene-alt-maleic anhydride)-b-poly(styrene) NPs, resulting in ∼50 nm particles with ∼ -30 mV surface charge. A peptide with high affinity for tartrate-resistant acid phosphatase (TRAP), a protein deposited by osteoclasts on bone resorptive surfaces, was introduced to the NP corona to achieve preferential delivery to fractured bone. Targeted NPs showed improved pharmacokinetic profiles with greater accumulation at fractured bone, accompanied by significant uptake in regenerative cell types (mesenchymal stem cells (MSCs) and osteoblasts). MSCs treated with drug-loaded NPs in vitro exhibited 2-fold greater ß-catenin signaling than free drug that was sustained for 5 days. To verify similar activity in vivo, TOPGAL reporter mice bearing fractures were treated with targeted GSK-3ß inhibitor-loaded NPs. Robust ß-galactosidase activity was observed in fracture callus and periosteum treated with targeted carriers versus controls, indicating potent ß-catenin activation during the healing process. Enhanced bone formation and microarchitecture were observed in mice treated with GSK-3ß inhibitor delivered via TRAP-binding peptide-targeted NPs. Specifically, increased bone bridging, ∼4-fold greater torsional rigidity, and greater volumes of newly deposited bone were observed 28 days after treatment, indicating expedited fracture healing.


Asunto(s)
Portadores de Fármacos/química , Curación de Fractura/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Nanopartículas/química , Péptidos/química , Inhibidores de Proteínas Quinasas/administración & dosificación , beta Catenina/agonistas , Animales , Huesos/efectos de los fármacos , Huesos/metabolismo , Huesos/patología , Células Cultivadas , Sistemas de Liberación de Medicamentos , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/uso terapéutico , beta Catenina/metabolismo
10.
ACS Nano ; 11(8): 7736-7746, 2017 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-28727410

RESUMEN

Organ development requires complex signaling by cells in different tissues. Epithelium and mesenchyme interactions are crucial for the development of skin, hair follicles, kidney, lungs, prostate, major glands, and teeth. Despite myriad literature on cell-cell interactions and ligand-receptor binding, the roles of extracellular vesicles in epithelium-mesenchyme interactions during organogenesis are poorly understood. Here, we discovered that ∼100 nm exosomes were secreted by the epithelium and mesenchyme of a developing tooth organ and diffused through the basement membrane. Exosomes were entocytosed by epithelium or mesenchyme cells with preference by reciprocal cells rather than self-uptake. Exosomes reciprocally evoked cell differentiation and matrix synthesis: epithelium exosomes induce mesenchyme cells to produce dentin sialoprotein and undergo mineralization, whereas mesenchyme exosomes induce epithelium cells to produce basement membrane components, ameloblastin and amelogenenin. Attenuated exosomal secretion by Rab27a/b knockdown or GW4869 disrupted the basement membrane and reduced enamel and dentin production in organ culture and reduced matrix synthesis and the size of the cervical loop, which harbors epithelium stem cells, in Rab27aash/ash mutant mice. We then profiled exosomal constituents including miRNAs and peptides and further crossed all epithelium exosomal miRNAs with literature-known miRNA Wnt regulators. Epithelium exosome-derived miR135a activated Wnt/ß-catenin signaling and escalated mesenchymal production of dentin matrix proteins, partially reversible by Antago-miR135a attenuation. Our results suggest that exosomes may mediate epithelium-mesenchyme crosstalk in organ development, suggesting that these vesicles and/or the molecular contents they are transporting may be interventional targets for treatment of diseases or regeneration of tissues.


Asunto(s)
Epitelio/metabolismo , Exosomas/metabolismo , Mesodermo/metabolismo , Animales , Western Blotting , Diferenciación Celular , Exosomas/genética , Técnica del Anticuerpo Fluorescente , Ratones , MicroARNs/genética , Microscopía Electrónica , Ratas , Ratas Sprague-Dawley , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo
11.
Bone Res ; 5: 17013, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28529816

RESUMEN

Stress during prenatal development is correlated with detrimental cognitive and behavioral outcomes in offspring. However, the long-term impact of prenatal stress (PS) and disrupted glucocorticoid signaling on bone mass and strength is not understood. In contrast, the detrimental effect of lead (Pb) on skeletal health is well documented. As stress and Pb act on common biological targets via glucocorticoid signaling pathways and co-occur in the environment, this study first sought to assess the combined effect of stress and Pb on bone quality in association with alterations in glucocorticoid signaling. Bone parameters were evaluated using microCT, histomorphometry, and strength determination in 8-month-old male mouse offspring subjected to PS on gestational days 16 and 17, lifetime Pb exposure (100 p.p.m. Pb in drinking water), or to both. Pb reduced trabecular bone mass and, when combined with PS, Pb unmasked an exaggerated decrement in bone mass and tensile strength. Next, to characterize a mechanism of glucocorticoid effect on bone, prednisolone was implanted subcutaneously (controlled-release pellet, 5 mg·kg-1 per day) in 5-month-old mice that decreased osteoblastic activity and increased sclerostin and leptin levels. Furthermore, the synthetic glucocorticoid dexamethasone alters the anabolic Wnt signaling pathway. The Wnt pathway inhibitor sclerostin has several glucocorticoid response elements, and dexamethasone administration to osteoblastic cells induces sclerostin expression. Dexamethasone treatment of isolated bone marrow cells decreased bone nodule formation, whereas removal of sclerostin protected against this decrement in mineralization. Collectively, these findings suggest that bone loss associated with steroid-induced osteoporosis is a consequence of sclerostin-mediated restriction of Wnt signaling, which may mechanistically facilitate glucocorticoid toxicity in bone.

12.
Nat Commun ; 7: 13073, 2016 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-27721375

RESUMEN

Tissue regeneration using stem cell-based transplantation faces many hurdles. Alternatively, therapeutically exploiting endogenous stem cells to regenerate injured or diseased tissue may circumvent these challenges. Here we show resident fibrocartilage stem cells (FCSCs) can be used to regenerate and repair cartilage. We identify FCSCs residing within the superficial zone niche in the temporomandibular joint (TMJ) condyle. A single FCSC spontaneously generates a cartilage anlage, remodels into bone and organizes a haematopoietic microenvironment. Wnt signals deplete the reservoir of FCSCs and cause cartilage degeneration. We also show that intra-articular treatment with the Wnt inhibitor sclerostin sustains the FCSC pool and regenerates cartilage in a TMJ injury model. We demonstrate the promise of exploiting resident FCSCs as a regenerative therapeutic strategy to substitute cell transplantation that could be beneficial for patients suffering from fibrocartilage injury and disease. These data prompt the examination of utilizing this strategy for other musculoskeletal tissues.


Asunto(s)
Fibrocartílago/citología , Regeneración , Trasplante de Células Madre , Células Madre/citología , Articulación Temporomandibular/patología , Articulación Temporomandibular/fisiopatología , Cicatrización de Heridas , Proteínas Adaptadoras Transductoras de Señales , Animales , Huesos/patología , Diferenciación Celular , Condrocitos/patología , Glicoproteínas/metabolismo , Homeostasis , Humanos , Péptidos y Proteínas de Señalización Intercelular , Masculino , Ratones Desnudos , Modelos Biológicos , Conejos , Ratas Sprague-Dawley , Nicho de Células Madre , Proteínas Wnt/metabolismo , Vía de Señalización Wnt
13.
Nat Commun ; 7: 10526, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26830436

RESUMEN

The suture mesenchyme serves as a growth centre for calvarial morphogenesis and has been postulated to act as the niche for skeletal stem cells. Aberrant gene regulation causes suture dysmorphogenesis resulting in craniosynostosis, one of the most common craniofacial deformities. Owing to various limitations, especially the lack of suture stem cell isolation, reconstruction of large craniofacial bone defects remains highly challenging. Here we provide the first evidence for an Axin2-expressing stem cell population with long-term self-renewing, clonal expanding and differentiating abilities during calvarial development and homeostastic maintenance. These cells, which reside in the suture midline, contribute directly to injury repair and skeletal regeneration in a cell autonomous fashion. Our findings demonstrate their true identity as skeletal stem cells with innate capacities to replace the damaged skeleton in cell-based therapy, and permit further elucidation of the stem cell-mediated craniofacial skeletogenesis, leading to revealing the complex nature of congenital disease and regenerative medicine.


Asunto(s)
Desarrollo Óseo/fisiología , Huesos Faciales/citología , Células Madre Mesenquimatosas/fisiología , Regeneración/fisiología , Cráneo/fisiología , Animales , Diferenciación Celular , Huesos Faciales/crecimiento & desarrollo , Trasplante de Células Madre Mesenquimatosas , Ratones
14.
Clin Chim Acta ; 455: 87-92, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26826396

RESUMEN

BACKGROUND: TGF-ß1 regulates bone metabolism and mediates bone turnover during postmenopause. Sclerostin negatively regulates Wnt signaling pathway and also has an important role in postmenopausal bone loss. Little is known about the relationship between serum TGF-ß1 and sclerostin during menopause. METHODS: We compared serum levels of TGF-ß1 and sclerostin in pre- and postmenopausal women and assessed the potential correlations of these levels with each other and with serum levels of bone turnover markers and bone mineral density. RESULTS: A total of 176 women (58 premenopausal, 62 early postmenopausal, and 56 late postmenopausal) were included in this study. Serum TGF-ß1 level was significantly higher in early postmenopausal women compared with premenopausal (32.0±7.19 vs. 26.55±6.67 ng/ml, p=0.01) and late postmenopausal (32.0±7.19 vs. 28.65±7.70 pg/ml, p=0.031) women, and no significant differences in serum sclerostin levels were observed among the 3 groups. There was a significant negative correlation between TGF-ß1 and sclerostin in early postmenopausal women, but not in other groups of women. Based on multiple regression analysis, only TGF-ß1 (ß=-0.362; p=0.007) was an independent predictor of sclerostin during early postmenopause. CONCLUSIONS: Our findings suggest that serum TGF-ß1 level increases during postmenopause and declines in old age. Sclerostin production is inhibited by TGF-ß1 during early postmenopause.


Asunto(s)
Proteínas Morfogenéticas Óseas/sangre , Posmenopausia , Factor de Crecimiento Transformador beta1/sangre , Proteínas Adaptadoras Transductoras de Señales , Estudios de Cohortes , Estudios Transversales , Femenino , Marcadores Genéticos , Humanos , Persona de Mediana Edad
15.
Toxicol Sci ; 149(2): 277-88, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26518054

RESUMEN

The heavy metal lead (Pb) has a deleterious effect on skeletal health. Because bone mass is maintained through a balance of bone formation and resorption, it is important to understand the effect of Pb levels on osteoblastic and osteoclastic activity. Pb exposure is associated with low bone mass in animal models and human populations; however, the correlation between Pb dosing and corresponding bone mass has been poorly explored. Thus, mice were exposed to increasing Pb and at higher levels (500 ppm), there was unexpectedly an increase in femur-tibial bone mass by 3 months of age. This is contrary to several studies alluded to earlier. Increased bone volume (BV) was accompanied by a significant increase in cortical thickness of the femur and trabecular bone that extended beyond the epiphyseal area into the marrow cavity. Subsequent evaluations revealed an increase in osteoclast numbers with high Pb exposure, but a deficiency in osteoclastic activity. These findings were substantiated by observed increases in levels of the resorption-altering hormones calcitonin and estrogen. In addition we found that pro-osteoclastic nuclear factor-kappa beta (NF-κB) pathway activity was dose dependently elevated with Pb, both in vivo and in vitro. However, the ability of osteoclasts to resorb bone was depressed in the presence of Pb in media and within test bone wafers. These findings indicate that exposure to high Pb levels disrupts early life bone accrual that may involve a disruption of osteoclast activity. This study accentuates the dose dependent variation in Pb exposure and consequent effects on skeletal health.


Asunto(s)
Densidad Ósea/efectos de los fármacos , Plomo/toxicidad , Osteoclastos/efectos de los fármacos , Adipocitos/efectos de los fármacos , Envejecimiento , Animales , Femenino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/fisiología , Osteoclastos/fisiología , Osteogénesis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Resistencia a la Tracción/efectos de los fármacos
16.
J Bone Miner Res ; 31(3): 549-59, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26363286

RESUMEN

WNT/ß-CATENIN signaling is involved in multiple aspects of skeletal development, including chondrocyte differentiation and maturation. Although the functions of ß-CATENIN in chondrocytes have been extensively investigated through gain-of-function and loss-of-function mouse models, the precise downstream effectors through which ß-CATENIN regulates these processes are not well defined. Here, we report that the matricellular protein, CCN1, is induced by WNT/ß-CATENIN signaling in chondrocytes. Specifically, we found that ß-CATENIN signaling promotes CCN1 expression in isolated primary sternal chondrocytes and both embryonic and postnatal cartilage. Additionally, we show that, in vitro, CCN1 overexpression promotes chondrocyte maturation, whereas inhibition of endogenous CCN1 function inhibits maturation. To explore the role of CCN1 on cartilage development and homeostasis in vivo, we generated a novel transgenic mouse model for conditional Ccn1 overexpression and show that cartilage-specific CCN1 overexpression leads to chondrodysplasia during development and cartilage degeneration in adult mice. Finally, we demonstrate that CCN1 expression increases in mouse knee joint tissues after meniscal/ligamentous injury (MLI) and in human cartilage after meniscal tear. Collectively, our data suggest that CCN1 is an important regulator of chondrocyte maturation during cartilage development and homeostasis.


Asunto(s)
Cartílago Articular/crecimiento & desarrollo , Diferenciación Celular , Condrocitos/metabolismo , Condrocitos/patología , Proteína 61 Rica en Cisteína/metabolismo , Animales , Animales Recién Nacidos , Apoptosis , Cartílago Articular/patología , Condrogénesis , Epífisis/patología , Menisco/patología , Ratones , Membrana Sinovial/patología , Vía de Señalización Wnt , beta Catenina/metabolismo
17.
J Biol Chem ; 290(29): 18216-18226, 2015 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-25975268

RESUMEN

Exposure to lead (Pb) from environmental sources remains an overlooked and serious public health risk. Starting in childhood, Pb in the skeleton can disrupt epiphyseal plate function, constrain the growth of long bones, and prevent attainment of a high peak bone mass, all of which will increase susceptibility to osteoporosis later in life. We hypothesize that the effects of Pb on bone mass, in part, come from depression of Wnt/ß-catenin signaling, a critical anabolic pathway for osteoblastic bone formation. In this study, we show that depression of Wnt signaling by Pb is due to increased sclerostin levels in vitro and in vivo. Downstream activation of the ß-catenin pathway using a pharmacological inhibitor of GSK-3ß ameliorates the Pb inhibition of Wnt signaling activity in the TOPGAL reporter mouse. The effect of Pb was determined to be dependent on sclerostin expression through use of the SOST gene knock-out mice, which are resistant to Pb-induced trabecular bone loss and maintain their mechanical bone strength. Moreover, isolated bone marrow cells from the sclerostin null mice show improved bone formation potential even after exposure to Pb. Also, our data suggest that the TGFß canonical signaling pathway is the mechanism by which Pb controls sclerostin production. Taken together these results support our hypothesis that the osteoporotic-like phenotype observed after Pb exposure is, in part, regulated through modulation of the Wnt/ß-catenin pathway.


Asunto(s)
Contaminantes Ambientales/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Plomo/toxicidad , Osteogénesis/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales , Animales , Huesos/anatomía & histología , Huesos/efectos de los fármacos , Huesos/metabolismo , Línea Celular , Células Cultivadas , Exposición a Riesgos Ambientales/efectos adversos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Glicoproteínas/genética , Péptidos y Proteínas de Señalización Intercelular , Masculino , Ratones , Ratones Noqueados , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , beta Catenina/metabolismo
18.
Environ Health Perspect ; 123(10): 935-43, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25861094

RESUMEN

BACKGROUND: Lead (Pb) exposure and obesity are co-occurring risk factors for decreased bone mass in the young, particularly in low socioeconomic communities. OBJECTIVES: The goal of this study was to determine whether the comorbidities of Pb exposure and high-fat diet-induced obesity amplify skeletal deficits independently associated with each of these risk factors, and to explore associated mechanisms of the observed deficiencies. METHODS: Five-week-old male C57BL/6J mice were placed on low-fat (10% kcal, LFD) or high-fat (60% kcal, HFD) diets for 12 weeks. Mice were exposed to lifetime Pb (50 ppm) through drinking water. RESULTS: HFD was associated with increased body mass and glucose intolerance. Both HFD and Pb increased fasting glucose and serum leptin levels. Pb and HFD each reduced trabecular bone quality and together had a further detrimental effect on these bone parameters. Mechanical bone properties of strength were depressed in Pb-exposed bones, but HFD had no significant effect. Both Pb and HFD altered progenitor cell differentiation, promoting osteoclastogenesis and increasing adipogenesis while suppressing osteoblastogenesis. In support of this lineage shift being mediated through altered Wnt signaling, Pb and non-esterified fatty acids in MC3T3 cells increased in vitro PPAR-γ activity and inhibited ß-catenin activity. Combining Pb and non-esterified fatty acids enhanced these effects. CONCLUSIONS: Pb and HFD produced selective deficits in bone accrual that were associated with alterations in progenitor cell activity that may involve reduced Wnt signaling. This study emphasizes the need to assess toxicants together with other risk factors relevant to human health and disease.


Asunto(s)
Desarrollo Óseo/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Contaminantes Ambientales/toxicidad , Plomo/toxicidad , Células Madre Mesenquimatosas/efectos de los fármacos , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/inducido químicamente , Vía de Señalización Wnt/efectos de los fármacos
19.
Bone ; 72: 92-100, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25446888

RESUMEN

PGE2 exerts anabolic and catabolic effects on bone through the discrete actions of four prostanoid receptors (EP1-4). We have previously demonstrated that loss EP1 accelerates fracture repair by enhancing bone formation. In the present study we defined the role of EP1 in bone maintenance and homeostasis during aging and in response to ovariectomy. The femur and L4 vertebrae of wild type (WT) and EP1(-/-) mice were examined at 2-months, 6-months, and 1-year of age, and in WT and EP1(-/-) mice following ovariectomy (OVX) or sham surgery. Bone volume fraction, trabecular architecture and mechanical properties were maintained during aging in EP1(-/-) mice to a greater degree than age-matched WT mice. Moreover, significant increases in bone formation rate (BFR) (+60%) and mineral apposition rate (MAR) (+50%) were observed in EP1(-/-), relative to WT, while no change in osteoclast number and osteoclast surface were observed. Following OVX, loss of EP1 was protective against bone loss in both femur and L4 vertebrae, with increased bone volume/total volume (BV/TV) (+32% in femur) and max load at failure (+10% in femur) relative to WT OVX, likely resulting from the increased bone formation rate that was observed in these mice. Taken together these studies identify inhibition of EP1 as a potential therapeutic approach to suppress bone loss in aged or post-menopausal patients.


Asunto(s)
Huesos/metabolismo , Subtipo EP1 de Receptores de Prostaglandina E/genética , Subtipo EP1 de Receptores de Prostaglandina E/metabolismo , Envejecimiento , Animales , Diferenciación Celular , Femenino , Homeostasis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Fluorescente , Osteoclastos/citología , Ovariectomía , Transducción de Señal , Estrés Mecánico , Microtomografía por Rayos X
20.
J Orthop Res ; 32(11): 1397-405, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25044211

RESUMEN

There is strong evidence in the clinical literature to suggest that elevated lead (Pb) exposure impairs fracture healing. Since Pb has been demonstrated to inhibit bone formation, and Wnt signaling is an important anabolic pathway in chondrocyte maturation and endochondral ossification, we investigated the impact of Wnt therapy on Pb-exposed mice undergoing bone repair in a mouse tibial fracture model. We established that tibial fracture calluses from Pb-treated mice were smaller and contained less mineralized tissue than vehicle controls. This resulted in the persistence of immature cartilage in the callus and decreased ß-catenin levels. Reduction of ß-catenin protein was concurrent with systemic elevation of LRP5/6 antagonists DKK1 and sclerostin in Pb-exposed mice throughout fracture healing. ß-catenin stimulation by the GSK3 inhibitor BIO reversed these molecular changes and restored the amount of mineralized callus. Overall, Pb is identified as a potent inhibitor of endochondral ossification in vivo with correlated effects on bone healing with noted deficits in ß-catenin signaling, suggesting the Wnt/ß-catenin as a pivotal pathway in the influence of Pb on fracture repair.


Asunto(s)
Curación de Fractura/efectos de los fármacos , Plomo/química , Células Madre Mesenquimatosas/citología , Fracturas de la Tibia/fisiopatología , beta Catenina/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales , Animales , Femenino , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glicoproteínas/metabolismo , Indoles/química , Péptidos y Proteínas de Señalización Intercelular , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoblastos/efectos de los fármacos , Osteogénesis , Oximas/química , Transducción de Señal , Fracturas de la Tibia/metabolismo , Factores de Tiempo , Proteínas Wnt/metabolismo , Cicatrización de Heridas , Microtomografía por Rayos X , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...