Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Cell Biochem ; 113(2): 640-9, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21956231

RESUMEN

Though extensive studies have been conducted, questions regarding the molecular effectors and pathways underlying the regulatory role of 1,25(OH)(2)D(3) in human osteoblasts other than cell differentiation and matrix protein production remain unanswered. This study aims to identify genes and pathways that are modulated by 1,25(OH)(2)D(3) treatment in human osteoblasts. Primary osteoblast cultures obtained from human bone tissue samples were treated with 1,25(OH)(2)D(3) (10(-7) M) for 24 h and their transcritptomes were profiled by microarray analysis using the Affymetrix GeneChip. Statistical analysis was conducted to identify genes whose expression is significantly modulated following 1,25(OH)(2)D(3) treatment. One hundred and fifty-eight genes were found to be differentially expressed. Of these, 136 were upregulated, indicating clear transcriptional activation by 1,25(OH)(2)D(3). Biostatistical evaluation of microarray data by Ingenuity Pathways Analysis (IPA) revealed a relevant modulation of genes involved in vitamin D metabolism (CYP24), immune functions (CD14), neurotransmitter transporters (SLC1A1, SLC22A3), and coagulation [thrombomodulin (THBD), tissue plasminogen activator (PLAT), endothelial protein C receptor (PROCR), thrombin receptor (F2R)]. We identified a restricted number of highly regulated genes and confirmed their differential expression by real-time quantitative PCR (RT qPCR). The present genome-wide microarray analysis on 1,25(OH)(2)D(3) -treated human osteoblasts reveals an interplay of critical regulatory and metabolic pathways and supports the hypothesis that 1,25(OH)(2)D(3) can modulate the coagulation process through osteoblasts, activates osteoclastogenesis through inflammation signaling, modulates the effects of monoamines by affecting their reuptake.


Asunto(s)
Calcitriol/farmacología , Regulación de la Expresión Génica , Osteoblastos/metabolismo , Vitaminas/farmacología , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Diferenciación Celular , Células Cultivadas , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Cultivo Primario de Células , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
2.
Blood ; 116(24): 5130-9, 2010 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-20847202

RESUMEN

Type I mucopolysaccharidosis (MPS I) is a lysosomal storage disorder caused by the deficiency of α-L-iduronidase, which results in glycosaminoglycan accumulation in tissues. Clinical manifestations include skeletal dysplasia, joint stiffness, visual and auditory defects, cardiac insufficiency, hepatosplenomegaly, and mental retardation (the last being present exclusively in the severe Hurler variant). The available treatments, enzyme-replacement therapy and hematopoietic stem cell (HSC) transplantation, can ameliorate most disease manifestations, but their outcome on skeletal and brain disease could be further improved. We demonstrate here that HSC gene therapy, based on lentiviral vectors, completely corrects disease manifestations in the mouse model. Of note, the therapeutic benefit provided by gene therapy on critical MPS I manifestations, such as neurologic and skeletal disease, greatly exceeds that exerted by HSC transplantation, the standard of care treatment for Hurler patients. Interestingly, therapeutic efficacy of HSC gene therapy is strictly dependent on the achievement of supranormal enzyme activity in the hematopoietic system of transplanted mice, which allows enzyme delivery to the brain and skeleton for disease correction. Overall, our data provide evidence of an efficacious treatment for MPS I Hurler patients, warranting future development toward clinical testing.


Asunto(s)
Terapia Genética/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Iduronidasa/administración & dosificación , Mucopolisacaridosis I/terapia , Animales , Huesos/efectos de los fármacos , Huesos/metabolismo , Huesos/patología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Vectores Genéticos , Iduronidasa/genética , Lentivirus/genética , Ratones , Ratones Noqueados , Mucopolisacaridosis I/patología , Fenotipo , Inducción de Remisión , Resultado del Tratamiento
3.
Amino Acids ; 43(4): 1751-9, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22407485

RESUMEN

Recent research suggests a role for ghrelin in the modulation of inflammatory disorders. However, the type of ghrelin receptor (GHS-R) involved in both the anti-inflammatory and anti-hyperalgesic actions of ghrelin remains to be characterized. In this study, we examined whether the inhibitory effect of ghrelin in the development of hyperalgesia and edema induced by intraplantar carrageenan administration depends on an interaction with GHS-R1a. Both central (1 nmol/rat, i.c.v.) and peripheral (40 nmol/kg, i.p.) administration of the selective GHS-R1a agonist EP1572 had no effect on carrageenan-induced hyperalgesia measured by Randall-Selitto test and paw edema. Furthermore, pre-treatment with the selective GHS-R1a antagonist, D-lys(3)-GHRP-6 (3 nmol/rat, i.c.v.) failed to prevent the anti-hyperalgesic and anti-inflammatory effects exerted by central ghrelin administration (1 nmol/rat), thus indicating that the type 1a GHS-R is not involved in these peptide activities. Accordingly, both central (1 and 2 nmol/rat, i.c.v.) and peripheral (40 and 80 nmol/kg, i.p.) administration of desacyl-ghrelin (DAG), which did not bind GHS-R1a, induced a significant reduction of the hyperalgesic and edematous activities of carrageenan. In conclusion, we have shown for the first time that DAG shares with ghrelin an inhibitory role in the development of hyperalgesia, as well as the paw edema induced by carrageenan and that a ghrelin receptor different from type 1a is involved in the anti-inflammatory activities of the peptide.


Asunto(s)
Ghrelina/farmacología , Inflamación/tratamiento farmacológico , Dolor/tratamiento farmacológico , Receptores de Ghrelina/agonistas , Receptores de Ghrelina/antagonistas & inhibidores , Animales , Carragenina , Edema/inducido químicamente , Edema/tratamiento farmacológico , Edema/metabolismo , Ghrelina/metabolismo , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Indoles , Inflamación/inducido químicamente , Inflamación/metabolismo , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Masculino , Oligopéptidos/farmacología , Dolor/inducido químicamente , Dolor/metabolismo , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Ghrelina/metabolismo , Triptófano/análogos & derivados
4.
Blood ; 114(15): 3216-26, 2009 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-19633200

RESUMEN

Adenosine deaminase (ADA) deficiency is a disorder of the purine metabolism leading to combined immunodeficiency and systemic alterations, including skeletal abnormalities. We report that ADA deficiency in mice causes a specific bone phenotype characterized by alterations of structural properties and impaired mechanical competence. These alterations are the combined result of an imbalanced receptor activator of nuclear factor-kappaB ligand (RANKL)/osteoprotegerin axis, causing decreased osteoclastogenesis and an intrinsic defect of osteoblast function with subsequent low bone formation. In vitro, osteoblasts lacking ADA displayed an altered transcriptional profile and growth reduction. Furthermore, the bone marrow microenvironment of ADA-deficient mice showed a reduced capacity to support in vitro and in vivo hematopoiesis. Treatment of ADA-deficient neonatal mice with enzyme replacement therapy, bone marrow transplantation, or gene therapy resulted in full recovery of the altered bone parameters. Remarkably, untreated ADA-severe combined immunodeficiency patients showed a similar imbalance in RANKL/osteoprotegerin levels alongside severe growth retardation. Gene therapy with ADA-transduced hematopoietic stem cells increased serum RANKL levels and children's growth. Our results indicate that the ADA metabolism represents a crucial modulatory factor of bone cell activities and remodeling.


Asunto(s)
Adenosina Desaminasa , Huesos/metabolismo , Trasplante de Células Madre Hematopoyéticas , Osteoblastos/metabolismo , Osteogénesis , Osteoprotegerina/sangre , Ligando RANK/sangre , Inmunodeficiencia Combinada Grave/sangre , Inmunodeficiencia Combinada Grave/terapia , Animales , Huesos/patología , Femenino , Terapia Genética , Hematopoyesis , Células Madre Hematopoyéticas/enzimología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Osteoprotegerina/genética , Ligando RANK/genética , Inmunodeficiencia Combinada Grave/patología , Trasplante Homólogo
5.
Blood ; 114(20): 4566-74, 2009 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-19773545

RESUMEN

The liver is the current site for pancreatic islet transplantation, but has many drawbacks due to immunologic and nonimmunologic factors. We asked whether pancreatic islets could be engrafted in the bone marrow (BM), an easily accessible and widely distributed transplant site that may lack the limitations seen in the liver. Syngeneic islets engrafted efficiently in the BM of C57BL/6 mice rendered diabetic by streptozocin treatment. For more than 1 year after transplantation, these animals showed parameters of glucose metabolism that were similar to those of nondiabetic mice. Islets in BM had a higher probability to reach euglycemia than islets in liver (2.4-fold increase, P = .02), showed a compact morphology with a conserved ratio between alpha and beta cells, and affected bone structure only very marginally. Islets in BM did not compromise hematopoietic activity, even when it was strongly induced in response to a BM aplasia-inducing infection with lymphocytic choriomeningitis virus. In conclusion, BM is an attractive and safe alternative site for pancreatic islet transplantation. The results of our study open a research line with potentially significant clinical impact, not only for the treatment of diabetes, but also for other diseases amenable to treatment with cellular transplantation.


Asunto(s)
Médula Ósea/cirugía , Diabetes Mellitus Experimental/cirugía , Trasplante de Islotes Pancreáticos/métodos , Islotes Pancreáticos , Animales , Glucemia , Supervivencia de Injerto , Inmunohistoquímica , Islotes Pancreáticos/anatomía & histología , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
6.
J Cell Physiol ; 225(3): 701-8, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20533307

RESUMEN

Transgenic mice over-expressing calcitonin gene-related peptide (CGRP) in osteoblasts have increased bone density due to increased bone formation, thus suggesting that CGRP plays a role in bone metabolism. In this study we determined the relationship between CGRP, the canonical Wnt signaling and apoptosis in human osteoblasts (hOBs) in consideration of the well-documented involvement of this pathway in bone cells. Primary cultures of hOBs were treated with CGRP 10(-8) M. Levels of ß-catenin, which is the cytoplasmic protein mediator of canonical Wnt signaling, and mRNA were determined. CGRP increases both the expression and the levels of cytoplasmic ß-catenin by binding to its receptor, as this effect is blocked by the antagonist CGRP(8-37). This facilitatory action on ß-catenin appears to be mediated by the inhibition of the enzyme GSK-3ß via protein kinase A (PKA) activation. GSK-3ß is a glycogen synthase kinase that, by phosphorylating ß-catenin, promotes its degradation by the proteosomal machinery. Moreover, the peptide is able to inhibit hOBs apoptosis stimulated by dexamethasone or by serum deprivation, possibly through the accumulation of ß-catenin, since the inhibitor of PKA activity H89 partially prevents the antiapoptotic effect of the peptide. In conclusion CGRP, released by nerve fibers, exerts its anabolic action on bone cells by stimulating canonical Wnt signaling and by inhibiting hOBs apoptosis, thus favoring local bone regeneration.


Asunto(s)
Apoptosis , Regeneración Ósea , Péptido Relacionado con Gen de Calcitonina/metabolismo , Osteoblastos/metabolismo , Fragmentos de Péptidos/metabolismo , beta Catenina/metabolismo , Transporte Activo de Núcleo Celular , Anciano , Apoptosis/efectos de los fármacos , Regeneración Ósea/efectos de los fármacos , Supervivencia Celular , Células Cultivadas , Colforsina/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Dexametasona/farmacología , Activadores de Enzimas/farmacología , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Isoquinolinas/farmacología , Persona de Mediana Edad , Osteoblastos/efectos de los fármacos , Osteoblastos/patología , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Estabilidad Proteica , ARN Mensajero/metabolismo , Transducción de Señal , Sulfonamidas/farmacología , Factores de Tiempo , Proteínas Wnt/metabolismo , beta Catenina/genética
7.
Neuroendocrinology ; 92(3): 189-97, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20805684

RESUMEN

BACKGROUND AND AIM: TLQP-21, a peptide derived from the vgf gene, has been reported to play a role in the regulation of rat gastric motility, but its influence on gastric mucosal integrity is unknown. EXPERIMENTAL APPROACH: We investigated the effects of central (0.8-8 nmol/rat) or peripheral (48-240 nmol/kg) TLQP-21 administration on ethanol- (EtOH, 50%, 1 ml/rat) induced gastric lesions in the rat. The mechanisms involved in such activity were also examined. RESULTS: Central TLQP-21 injection dose-dependently reduced EtOH-induced gastric lesions (ED(50) = 3.16 nmol), while peripheral TLQP-21 administration had no effect. The TLQP-21 gastroprotective effect against EtOH injury was accompanied by a significant increase in gastric prostaglandin E(2) (PGE(2)) production linked to an increase in constitutive cyclooxygenase (COX) expression. The nitric oxide (NO) synthase inhibitor L-NAME (70 mg/kg, s.c.), the nonselective COX inhibitor indomethacin (10 mg/kg, orally) and capsaicin denervation removed TLQP-21 gastroprotection. CONCLUSIONS: This study shows for the first time that central TLQP-21 exerts a protective action on the gastric mucosa exposed to the noxious agent EtOH. TLQP-21 gastroprotection is mediated by constitutive-derived NO and PGE(2), and requires the integrity of sensory nerve fibers.


Asunto(s)
Mucosa Gástrica/efectos de los fármacos , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Úlcera Gástrica/inducido químicamente , Úlcera Gástrica/prevención & control , Animales , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Etanol/farmacología , Mucosa Gástrica/inervación , Mucosa Gástrica/patología , Técnicas para Inmunoenzimas , Indometacina/farmacología , Modelos Lineales , Masculino , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Prostaglandina-Endoperóxido Sintasas/genética , Prostaglandina-Endoperóxido Sintasas/metabolismo , ARN Mensajero , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Úlcera Gástrica/patología
8.
Biochem Biophys Res Commun ; 371(1): 94-8, 2008 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-18413146

RESUMEN

We have previously shown that the biphenylcarboxylic acid butanediol ester (ABD56) inhibits osteoclast formation and activity in vitro and in vivo. However, the mechanism of action of this compound is unknown. ABD56 inhibited osteoclast formation and caused osteoclast apoptosis, but had no effects on osteoblasts or macrophages. As the NFkappaB and MAPK pathways are essential for osteoclast formation and survival, we studied the effects of ABD56 on these pathways. ABD56 caused phosphorylation of p38, JNK and nuclear translocation of c-jun in osteoclasts. ABD56-induced apoptosis was prevented by the caspase inhibitor zVAD-fmk but was not prevented by the p38- or JNK-inhibitors. ABD56 completely abolished RANKL-induced IkappaB and ERK1/2 phosphorylation. Increasing the amount of RANKL partially rescued ABD56-induced apoptosis, indicating that the apoptosis is most probably due to the inhibition of survival signals such as ERK and NFkappaB, rather than activation of the p38 or Jnk MAPK pathways.


Asunto(s)
Apoptosis , Benzoatos/farmacología , Compuestos de Bifenilo/farmacología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , FN-kappa B/antagonistas & inhibidores , Osteoclastos/efectos de los fármacos , Clorometilcetonas de Aminoácidos/farmacología , Animales , Apoptosis/efectos de los fármacos , Inhibidores de Caspasas , Células Cultivadas , Inhibidores de Cisteína Proteinasa/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ratones , FN-kappa B/metabolismo , Osteoclastos/metabolismo , Fosforilación , Ligando RANK/antagonistas & inhibidores , Ligando RANK/farmacología , Conejos , Transducción de Señal/efectos de los fármacos
9.
J Bone Miner Res ; 22(9): 1387-96, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17539736

RESUMEN

UNLABELLED: The uPAR and its ligand uPA are expressed by both osteoblasts and osteoclasts. Their function in bone remodeling is unknown. We report that uPAR-lacking mice display increased BMD, increased osteogenic potential of osteoblasts, decreased osteoclasts formation, and altered cytoskeletal reorganization in mature osteoclasts. INTRODUCTION: Urokinase receptor (uPAR) is actively involved in the regulation of important cell functions, such as proliferation, adhesion, and migration. It was previously shown that the major players in bone remodeling, osteoblasts and osteoclasts, express uPAR and produce urokinase (uPA). The purpose of this study was to investigate the role of uPAR in bone remodeling. MATERIALS AND METHODS: In vivo studies were performed in uPAR knockout (KO) and wildtype (WT) mice on a C57Bl6/SV129 (75:25) background. Bone mass was analyzed by pQCT. Excised tibias were subjected to mechanical tests. UPAR KO calvaria osteoblasts were characterized by proliferation assays, RT-PCR for important proteins secreted during differentiation, and immunoblot for activator protein 1 (AP-1) family members. In vitro osteoclast formation was tested with uPAR KO bone marrow monocytes in the presence of macrophage-colony stimulating factor (M-CSF) and RANKL. Phalloidin staining in osteoclasts served to study actin ring and podosome formation. RESULTS: pQCT revealed increased bone mass in uPAR-null mice. Mechanical tests showed reduced load-sustaining capability in uPAR KO tibias. uPAR KO osteoblasts showed a proliferative advantage with no difference in apoptosis, higher matrix mineralization, and earlier appearance of alkaline phosphatase (ALP). Surface RANKL expression at different stages of differentiation was not altered. AP-1 components, such as JunB and Fra-1, were upregulated in uPAR KO osteoblasts, along with other osteoblasts markers. On the resorptive side, the number of osteoclasts formed in vitro from uPAR KO monocytes was decreased. Podosome imaging in uPAR KO osteoclasts revealed a defect in actin ring formation. CONCLUSIONS: The defective proliferation and differentiation of bone cells, coincident with both aberrant expression of transcription factors and cytoskeletal organization, are typical uPAR-dependent molecular phenotypes, and we have now shown their function in osteoblasts and osteoclasts function in vivo.


Asunto(s)
Huesos/fisiología , Homeostasis/fisiología , Osteoblastos/citología , Osteoclastos/citología , Receptores de Superficie Celular/fisiología , Animales , Secuencia de Bases , Huesos/citología , Huesos/diagnóstico por imagen , Cartilla de ADN , Ratones , Ratones Noqueados , Tamaño de los Órganos , Receptores de Superficie Celular/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tomografía Computarizada por Rayos X
10.
Biomed Res Int ; 2016: 8169614, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27999816

RESUMEN

Wnt signaling, a major regulator of bone formation and homeostasis, might be involved in the bone loss of osteoporotic patients and the consequent impaired response to fracture. Therefore we analyzed Wnt-related, osteogenic, and adipogenic genes in bone tissue of elderly postmenopausal women undergoing hip replacement for either femoral fracture or osteoarthritis. Bone specimens derived from the intertrochanteric region of the femurs of 25 women with fracture (F) and 29 with osteoarthritis without fracture (OA) were analyzed. Specific miRNAs were analyzed in bone and in matched blood samples. RUNX2, BGP, and OPG showed lower expression in F than in OA samples, while OSX, OPN, BSP, and RANKL were not different. Inhibitory genes of Wnt pathway were lower in F versus OA. ß-Catenin protein levels were higher in F versus OA, whereas its cotranscriptional regulator (Lef1) was lower in F group. miR-204, which targets RUNX2, and miR-130a, which inhibits PPARγ, were lower and higher, respectively, in F versus OA serum samples. The present study showed an inefficient Wnt signal transduction in F group despite higher ß-catenin protein levels, consistent with the expected overall postfracture systemic activation towards osteogenesis. This transcriptional inefficiency could contribute to the osteoporotic bone fragility.


Asunto(s)
Fracturas del Fémur/sangre , Posmenopausia/sangre , Vía de Señalización Wnt , Anciano , Anciano de 80 o más Años , Subunidad alfa 1 del Factor de Unión al Sitio Principal/sangre , Femenino , Fracturas del Fémur/patología , Humanos , MicroARNs/sangre , Osteoartritis/sangre , Osteoartritis/patología , Osteoprotegerina/sangre , Ligando RANK/sangre , beta Catenina/sangre
11.
Int J Endocrinol ; 2015: 547473, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25866509

RESUMEN

Ghrelin, by binding growth hormone secretagogue receptor (GHS-R), promotes osteoblast proliferation but the signaling mechanism of GHS-R on these cells remains unclear. Since canonical Wnt/ß-catenin pathway is critically associated with bone homeostasis, we investigated its involvement in mediating ghrelin effects in osteoblasts and in osteoblast-osteoclast cross talk. Ghrelin (10(-10)M) significantly increased ß-catenin levels in rat osteoblasts (rOB). This stimulatory action on ß-catenin involves a specific interaction with GHS-R1a, as it is prevented by the selective GHS-R1a antagonist, D-Lys(3)-GHRP-6 (10(-7)M). The effect of ghrelin on ß-catenin involves the phosphorylation and inactivation of GSK-3ß via protein kinase A (PKA). Inhibition of PKA activity reduces the facilitatory action of ghrelin on ß-catenin stabilization. Ghrelin treatment of rOB significantly increases the expression of osteoprotegerin (OPG), which plays an important role in the regulation of osteoclastogenesis, and this effect is blocked by D-Lys(3)-GHRP-6. Furthermore, ghrelin reduced RANKL/OPG ratio thus contrasting osteoclastogenesis. Accordingly, conditioned media from rOB treated with ghrelin decreased the number of multinucleated TRAcP+ cells as compared with the conditioned media from untreated-control rOB. Our data suggest new roles for ghrelin in modulating bone homeostasis via a specific interaction with GHSR-1a in osteoblasts with subsequent enhancement of both ß-catenin levels and OPG expression.

12.
Endocrine ; 44(3): 790-802, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23619963

RESUMEN

Several studies support a serotonin role in the physiological control of bone mass. However, whether serotonin (5-HT) is involved in bone loss due to reduced mechanical stress or unloading is unknown. We investigated the effects of reduced 5-HT tone, induced by tryptophan-free diet, in movement-restraint osteopenia induced by housing mature rats, acclimatised in single cages with a floor area of 1,500 cm(2), in smaller size single cages where their motor activity was reduced. Tryptophan-deficiency significantly worsened movement-restraint-induced bone loss in both femoral metaphysis and diaphysis (DXA analysis) but not at lumbar vertebrae and impaired the mechanical properties of the femur by significantly reducing both cortical thickness and strength strain index (pQCT analysis). Such effects resulted from an impairment of bone turnover with bone resorption exceeding bone formation. Tryptophan-supplemented diet reversed the worsening effects of tryptophan-deficiency on movement-restraint osteopenia. The improvements of both bone mass and strength were associated with an increase of serum osteocalcin and IGF-I, markers of osteoblast activity. In vitro studies in primary cultures of rat osteoblasts suggest that the anabolic action of 5-HT involves the activation of the Wnt/ß-catenin pathway. Serotonin significantly increased the cytoplasmatic ß-catenin protein levels by the inhibition of the enzyme glycogen synthase kinase-3ß, that by phosphorylating ß-catenin promotes its degradation. Our data support a role for 5-HT in the anabolic response of the appendicular skeleton to mechanical loading. We suggest that serotonin might stimulate canonical Wnt/ß-catenin-dependent bone formation to occur.


Asunto(s)
Huesos/metabolismo , Dieta , Actividad Motora/fisiología , Serotonina/metabolismo , Triptófano/metabolismo , Animales , Densidad Ósea/fisiología , Células Cultivadas , Masculino , Osteoblastos/citología , Osteoblastos/metabolismo , Ratas , Ratas Sprague-Dawley
13.
Bone ; 55(1): 84-92, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23567159

RESUMEN

Recent evidence demonstrated an interplay between estrogens and growth hormone (GH) at cellular level. To investigate the possible mechanism/s involved, we studied the effect of 17ß-estradiol (E2) on GH signaling pathways in primary culture of human osteoblasts (hOBs). Exposure of hOBs to E2 (10(-8) M) 60 min before GH (5 ng/ml) significantly increased phosphorylated STAT5 (P-STAT5) levels compared with GH alone. E2 per se had no effect on P-STAT5. E2-enhanced GH signaling was effective in increasing osteopontin, bone-sialoprotein, and IGF II mRNA expression to a greater extent than GH alone. We then studied the effect of E2 on the protein levels of the negative regulator of GH signaling, suppressor of cytokine signaling-2 (SOCS2). E2 (10(-11) M-10(-7) M) reduced dose-dependently SOCS2 protein levels without modifying its mRNA expression. The silencing of SOCS2 gene prevented E2 positive effect on GH induced P-STAT5 and on GH induced bone-sialoprotein and osteopontin mRNA expression. Treatment with the inhibitor of DNA-dependent RNA synthesis, actinomycin-D, did not prevent E2 induced decrease of SOCS2, thus suggesting a non-genomic effect. E2 promoted an increase in SOCS2 ubiquitination. To determine if increased ubiquitination of SOCS2 by E2 led to degradation by proteasome, hOBs were pretreated with the proteasome inhibitor MG132 (5 µM) which blocked E2 reduction of SOCS2. These findings demonstrate for the first time that E2 can amplify GH intracellular signaling in hOBs with an essential role played by the reduction of the SOCS2 mediated feedback loop.


Asunto(s)
Estradiol/farmacología , Retroalimentación Fisiológica/efectos de los fármacos , Hormona de Crecimiento Humana/metabolismo , Osteoblastos/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Anciano , Anciano de 80 o más Años , Dactinomicina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , Sialoproteína de Unión a Integrina/genética , Sialoproteína de Unión a Integrina/metabolismo , Leupeptinas/farmacología , Osteoblastos/efectos de los fármacos , Osteopontina/genética , Osteopontina/metabolismo , Fosforilación/efectos de los fármacos , Proteolisis/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal/genética , Proteína 1 Supresora de la Señalización de Citocinas , Proteína 3 Supresora de la Señalización de Citocinas , Transfección , Ubiquitinación/efectos de los fármacos
14.
Peptides ; 30(12): 2233-41, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19682523

RESUMEN

Casein phosphopeptides (CPPs), originating by in vitro and/or in vivo casein digestion, are characterized by the ability to complex and solubilize calcium ions preventing their precipitation. Previous works demonstrated that CPPs improve calcium uptake by human differentiated intestinal tumor cell lines, are able to re-mineralize carious lesions in a dental enamel, and, as components of a diet, affect bone weight and calcium content in rats. The aim of the present study was to evaluate if CPPs can directly modulate bone cells activity and mineralization. Primary human osteoblast-like cells were established in culture from trabecular bone samples obtained from waste materials during orthopedic surgery. Commercial mixtures of bovine casein phosphopeptides were used. The CPP dependent intracellular calcium rises were monitored at the single cell level through fura2-fluorescence assays. Results show that CPPs: (i) stimulate calcium uptake by primary human osteoblast-like cells; (ii) increase the expression and activity of alkaline phosphatase, a marker of human osteoblast differentiation; (iii) affect the cell proliferation rate and the apoptotic level; (iv) enhance nodule formation by human SaOS-2. Taken together these results confirm the possibility that CPPs play a role as modulator of bone cell activity, probably sustained by their ability as calcium carriers. Although the exact mechanism by which CPPs act remains not completely clarified, they can be considered as potential anabolic factors for bone tissue engineering.


Asunto(s)
Calcio/metabolismo , Caseínas/metabolismo , Diferenciación Celular/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Fosfopéptidos/metabolismo , Fosfopéptidos/farmacología , Fosfatasa Alcalina/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Humanos , L-Lactato Deshidrogenasa/metabolismo , Osteoblastos/citología , Fosfopéptidos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA