Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Ann Oncol ; 29(12): 2334-2340, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30307466

RESUMEN

Background: Palbociclib is a CDK4/6 inhibitor with demonstrated efficacy and safety in combination with endocrine therapy in advanced luminal breast cancer (LBC). We evaluated the respective efficacy and safety of chemotherapy and letrozole-palbociclib (LETPAL) combination as neoadjuvant treatment in patients with high-risk LBC. Patients and methods: NeoPAL (UCBG10/4, NCT02400567) is a randomised, parallel, non-comparative phase II study. Patients with ER-positive, HER2-negative, Prosigna®-defined luminal B, or luminal A and node-positive, stage II-III breast cancer, not candidate for breast-conserving surgery, were randomly assigned to either letrozole (2.5 mg daily) and palbociclib (125 mg daily, 3 weeks/4) during 19 weeks, or to FEC100 (5FU 500 mg/m2, epirubicin 100 mg/m2, cyclophosphamide 500 mg/m2)×3 21-day courses followed by docetaxel 100 mg/m2×3 21-day courses. Primary end point was residual cancer burden (RCB 0-I rate). Secondary end points included clinical response, proliferation-based markers, and safety. Results: Overall, 106 patients were randomised [median Prosigna® ROR Score 71 (22-93)]. RCB 0-I was observed in four and eight patients in LETPAL [7.7% (95% CI 0.4-14.9)] and chemotherapy [15.7% (95% CI 5.7-25.7)] arms, respectively. Pathological complete response rates were 3.8% and 5.9%. Clinical response (75%) and breast-conserving surgery rates (69%) were similar in both arms. Preoperative Endocrine Prognostic Index 0 scores (breast cancer-specific survival) were observed in 17.6% and 8.0% of patients in LETPAL and chemotherapy arms, respectively. Safety profile was as expected, with 2 versus 17 serious adverse events (including 11 grade 4 serious AEs in the chemotherapy arm). Conclusion: LETPAL combination was associated with poor pathological response but encouraging clinical and biomarker responses in Prosigna®-defined high-risk LBC. Contemporary chemotherapy regimen was associated with poor pathological and biomarker responses, with a much less favourable safety profile. LETPAL combination might represent an alternative to chemotherapy in early high-risk LBC. Clinical Trial Number: NCT02400567.


Asunto(s)
Antineoplásicos Hormonales/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias de la Mama/terapia , Letrozol/administración & dosificación , Piperazinas/administración & dosificación , Inhibidores de Proteínas Quinasas/administración & dosificación , Piridinas/administración & dosificación , Anciano , Antineoplásicos Hormonales/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/genética , Mama/patología , Mama/cirugía , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Supervivencia sin Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Letrozol/efectos adversos , Mastectomía Segmentaria , Persona de Mediana Edad , Terapia Neoadyuvante/métodos , Estadificación de Neoplasias , Selección de Paciente , Piperazinas/efectos adversos , Pronóstico , Inhibidores de Proteínas Quinasas/efectos adversos , Piridinas/efectos adversos
2.
Ann Oncol ; 28(1): 103-109, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28177480

RESUMEN

Background: We present a pooled analysis of predictive and prognostic values of circulating tumour cells (CTC) and circulating endothelial cells (CEC) in two prospective trials of patients with inflammatory breast cancer (IBC) treated with neoadjuvant chemotherapy combined with neoadjuvant and adjuvant bevacizumab. Patients and methods: Nonmetastatic T4d patients were enrolled in two phase II multicentre trials, evaluating bevacizumab in combination with sequential neoadjuvant chemotherapy of four cycles of FEC followed by four cycles of docetaxel in HER2-negative tumour (BEVERLY-1) or docetaxel and trastuzumab in HER2-positive tumour (BEVERLY-2). CTC and CEC were detected in 7.5 and 4 ml of blood, respectively, with the CellSearch System. Results: From October 2008 to September 2010, 152 patients were included and 137 were evaluable for CTC and CEC. At baseline, 55 patients had detectable CTC (39%). After four cycles of chemotherapy, a dramatic drop in CTC to a rate of 9% was observed (P < 0.01). Pathological complete response (pCR) rate was 40%. No correlation was found between CTC or CEC levels and pCR rate. Median follow-up was 43 months. CTC detection (≥1 CTC/7.5 ml) at baseline was associated with shorter 3-year disease-free survival (39% versus 70% for patients without CTC, P < 0.01, HR 2.80) and shorter 3-year overall survival (OS) (P < 0.01). In multivariate analysis, independent prognostic parameters for shorter survival were absence of hormonal receptors, no pCR and CTC detection at baseline. CEC level at baseline or variations during treatment had no prognostic value. Conclusion: In this pooled analysis of two prospective trials in nonmetastatic IBC, detection rate of CTC was 39% with a strong and independent prognostic value for survival. Combination of pCR after neoadjuvant treatment with no CTC detection at baseline isolated a subgroup of IBC with excellent OS (94% 3-year OS), suggesting that CTC count could be part of IBC stratification in prospective trials.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Inflamatorias de la Mama/tratamiento farmacológico , Neoplasias Inflamatorias de la Mama/patología , Células Neoplásicas Circulantes/efectos de los fármacos , Células Neoplásicas Circulantes/patología , Adulto , Anciano , Bevacizumab/administración & dosificación , Quimioterapia Adyuvante , Ciclofosfamida/administración & dosificación , Supervivencia sin Enfermedad , Docetaxel , Epirrubicina/administración & dosificación , Femenino , Humanos , Neoplasias Inflamatorias de la Mama/sangre , Neoplasias Inflamatorias de la Mama/cirugía , Persona de Mediana Edad , Terapia Neoadyuvante , Pronóstico , Receptor ErbB-2/metabolismo , Taxoides/administración & dosificación , Trastuzumab/administración & dosificación , Adulto Joven
3.
Ann Oncol ; 26(8): 1692-7, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25943350

RESUMEN

BACKGROUND: Inflammatory breast cancer (IBC) is a rare and aggressive disease requiring a multimodal treatment. We evaluated the benefit of adding docetaxel-5-fluorouracil (D-5FU) regimen after preoperative dose-intense (DI) epirubicin-cyclophosphamide (EC) and locoregional treatment in IBC patients. PATIENTS AND METHODS: PEGASE 07 was a national randomized phase III open-label study involving 14 hospitals in France. Women with nonmetastatic IBC were eligible and randomly assigned to receive either four cycles of DI EC (E 150 mg/m(2) and C 4000 mg/m(2) every 3 weeks with repeated hematopoietic stem cell support), then mastectomy with axillary lymph node dissection, and radiotherapy (arm A) or the same treatment followed by four cycles of D-5FU (D 85 mg/m(2), day 1 and 5FU 750 mg/m(2)/day continuous infusion, days 1-5 every 3 weeks) administered postradiotherapy (arm B). Patients with hormone receptor-positive tumors received hormonal therapy. Disease-free survival (DFS) was the primary end point. Secondary end points included tolerance, pathological complete response (pCR) rate, and overall survival (OS). RESULTS: Between January 2001 and May 2005, 174 patients were enrolled and treated (87 in each arm). Median follow-up was similar in both arms: 59.6 months [95% confidence interval (CI) 58.4-60.3] in arm A and 60.5 months (95% CI 58.3-61.4) in arm B. The estimated 5-year DFS rates were not different: 55% (95% CI 43.9-64.7) in arm A and 55.5% (95% CI 44.3-65.3) in arm B [hazard ratio (HR) = 0.94 (0.61-1.48); P = 0.81]. Identical results were observed for 5-year OS: 70.2% (95% CI 59.1-78.8) in arm A and 70% (95% CI 58.8-78.7) in arm B [HR = 0.93 (0.55-1.60); P = 0.814]. Following DI EC induction, in-breast and global (breast plus nodes) pCR were 28.9% and 20.1%, respectively. Estrogen receptor and pCR status were independently associated with survival. CONCLUSION: The addition of D-5FU after preoperative DI EC and standard local therapy did not improve DFS in IBC. CLINICAL TRIAL NUMBER: ClinicalTrials.gov identifier: NCT02324088.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma Ductal de Mama/tratamiento farmacológico , Carcinoma Lobular/tratamiento farmacológico , Neoplasias Inflamatorias de la Mama/tratamiento farmacológico , Mastectomía , Terapia Neoadyuvante/métodos , Adulto , Antineoplásicos Hormonales/administración & dosificación , Axila , Quimioradioterapia Adyuvante/métodos , Quimioterapia Adyuvante/métodos , Ciclofosfamida/administración & dosificación , Supervivencia sin Enfermedad , Docetaxel , Epirrubicina/administración & dosificación , Femenino , Fluorouracilo/administración & dosificación , Humanos , Neoplasias Inflamatorias de la Mama/metabolismo , Escisión del Ganglio Linfático , Persona de Mediana Edad , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Taxoides/administración & dosificación
4.
ESMO Open ; 9(5): 103443, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38692082

RESUMEN

BACKGROUND: The purpose of this study was to evaluate the prognostic value of the multigene EndoPredict test in prospectively collected data of patients screened for the randomized, double-blind, phase III UNIRAD trial, which evaluated the addition of everolimus to adjuvant endocrine therapy in high-risk, hormone receptor-positive, human epidermal growth factor receptor 2 (HER2)-negative early breast cancer. PATIENTS AND METHODS: Patients were classified into low or high risk according to the EPclin score, consisting of a 12-gene molecular score combined with tumor size and nodal status. Association of the EPclin score with disease-free survival (DFS) and distant metastasis-free survival (DMFS) was evaluated using Kaplan-Meier estimates. The independent prognostic added value of EPclin score was tested in a multivariate Cox model after adjusting on tumor characteristics. RESULTS: EndoPredict test results were available for 768 patients: 663 patients classified as EPclin high risk (EPCH) and 105 patients as EPclin low risk (EPCL). Median follow-up was 70 months (range 1-172 months). For the 429 EPCH randomized patients, there was no significant difference in DFS between treatment arms. The 60-month relapse rate for patients in the EPCL and EPCH groups was 0% and 7%, respectively. Hazard ratio (HR) supposing continuous EPclin score was 1.87 [95% confidence interval (CI) 1.4-2.5, P < 0.0001]. This prognostic effect remained significant when assessed in a Cox model adjusting on tumor size, number of positive nodes and tumor grade (HR 1.52, 95% CI 1.09-2.13, P = 0.0141). The 60-month DMFS for patients in the EPCL and EPCH groups was 100% and 94%, respectively (adjusted HR 8.10, 95% CI 1.1-59.1, P < 0.0001). CONCLUSIONS: The results confirm the value of EPclin score as an independent prognostic parameter in node-positive, hormone receptor-positive, HER2-negative early breast cancer patients receiving standard adjuvant treatment. EPclin score can be used to identify patients at higher risk of recurrence who may warrant additional systemic treatments.


Asunto(s)
Neoplasias de la Mama , Receptor ErbB-2 , Humanos , Femenino , Neoplasias de la Mama/patología , Neoplasias de la Mama/tratamiento farmacológico , Persona de Mediana Edad , Pronóstico , Receptor ErbB-2/metabolismo , Método Doble Ciego , Anciano , Adulto , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Everolimus/uso terapéutico , Everolimus/farmacología , Supervivencia sin Enfermedad , Biomarcadores de Tumor/metabolismo
5.
J Clin Invest ; 101(6): 1310-7, 1998 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-9502772

RESUMEN

Apert syndrome, associated with fibroblast growth factor receptor (FGFR) 2 mutations, is characterized by premature fusion of cranial sutures. We analyzed proliferation and differentiation of calvaria cells derived from Apert infants and fetuses with FGFR-2 mutations. Histological analysis revealed premature ossification, increased extent of subperiosteal bone formation, and alkaline phosphatase- positive preosteoblastic cells in Apert fetal calvaria compared with age-matched controls. Preosteoblastic calvaria cells isolated from Apert infants and fetuses showed normal cell growth in basal conditions or in response to exogenous FGF-2. In contrast, the number of alkaline phosphatase- positive calvaria cells was fourfold higher than normal in mutant fetal calvaria cells with the most frequent Apert FGFR-2 mutation (Ser252Trp), suggesting increased maturation rate of cells in the osteoblastic lineage. Biochemical and Northern blot analyses also showed that the expression of alkaline phosphatase and type 1 collagen were 2-10-fold greater than normal in mutant fetal calvaria cells. The in vitro production of mineralized matrix formed by immortalized mutant fetal calvaria cells cultured in aggregates was also increased markedly compared with control immortalized fetal calvaria cells. The results show that Apert FGFR-2 mutations lead to an increase in the number of precursor cells that enter the osteogenic pathway, leading ultimately to increased subperiosteal bone matrix formation and premature calvaria ossification during fetal development, which establishes a connection between the altered genotype and cellular phenotype in Apert syndromic craniosynostosis.


Asunto(s)
Acrocefalosindactilia/genética , Acrocefalosindactilia/metabolismo , Matriz Ósea/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Cráneo/metabolismo , Fosfatasa Alcalina/metabolismo , Sustitución de Aminoácidos , Northern Blotting , Diferenciación Celular/genética , División Celular/genética , Células Cultivadas , Colágeno/metabolismo , Feto/citología , Feto/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Expresión Génica , Histocitoquímica , Humanos , Lactante , Nitrofenoles/metabolismo , Osteogénesis/genética , Mutación Puntual , Serina/genética , Cráneo/citología , Cráneo/crecimiento & desarrollo , Triptófano/genética
7.
J Bone Miner Res ; 16(4): 705-12, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11315998

RESUMEN

Apert (Ap) syndrome is a craniofacial malformation characterized by premature fusion of cranial sutures (craniosynostosis). We previously showed that the Ser252Trp fibroblast growth factor receptor 2 (FGFR-2) mutation in Ap syndrome increases osteoblast differentiation and subperiosteal bone matrix formation, leading to premature calvaria ossification. In this study, we used the emerging technology of complementary DNA (cDNA) microarray to identify genes that are involved in osteoblast abnormalities induced by the Ser252Trp FGFR-2 mutation. To identify the signaling pathways involved in this syndrome, we used radioactively labeled cDNAs derived from two sources of cellular messenger RNAs (mRNAs) for hybridization: control (Co) and mutant Ap immortalized osteoblastic cells. Among genes that were differentially expressed, protein kinase Ca (PKC-alpha), interleukin-1alpha (IL-1alpha), and the small guanosine-5'-triphosphatase (GTPase) RhoA were increased in FGFR-2 mutant Ap cells compared with Co cells. The validity of the hybridization array was confirmed by Northern blot analysis using mRNAs derived from different cultures. Furthermore, immunochemical and Western blot analyses showed that mutant Ap cells displayed increased PKC-alpha, IL-1alpha, and RhoA protein levels compared with Co cells. Treatment of Co and Ap cells with the PKC inhibitor calphostin C decreased IL-1alpha and RhoA mRNA and protein levels in Ap cells, indicating that PKC is upstream of IL-1alpha and RhoA. Moreover, SB203580, a specific inhibitor of p38 mitogen-activated protein kinase (MAPK), and PD-98059, a specific inhibitor of MAPK kinase (MEKK), also reduced IL-1alpha and RhoA expression in Ap cells. These data show that the Ser252Trp FGFR-2 mutation in Ap syndrome induces constitutive overexpression of PKC-alpha, IL-1alpha, and small GTPase RhoA, suggesting a role for these effectors in osteoblast alterations induced by the mutation. The cDNA microarray technology appears to be a useful tool to gain information on abnormal gene expression and molecular pathways induced by genetic mutations in bone cells.


Asunto(s)
Acrocefalosindactilia/genética , Sustitución de Aminoácidos , Regulación de la Expresión Génica , Interleucina-1/biosíntesis , Isoenzimas/biosíntesis , Quinasa 1 de Quinasa de Quinasa MAP , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteoblastos/metabolismo , Mutación Puntual , Proteína Quinasa C/biosíntesis , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Factores de Crecimiento de Fibroblastos/genética , Proteína de Unión al GTP rhoA/biosíntesis , Acrocefalosindactilia/embriología , Acrocefalosindactilia/metabolismo , Acrocefalosindactilia/patología , Línea Celular Transformada/metabolismo , ADN Complementario/genética , Inducción Enzimática , Inhibidores Enzimáticos/farmacología , Feto , Flavonoides/farmacología , Humanos , Imidazoles/farmacología , Interleucina-1/genética , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Naftalenos/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , Proteína Quinasa C-alfa , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Piridinas/farmacología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Cráneo/embriología , Cráneo/patología , Técnica de Sustracción , Proteínas Quinasas p38 Activadas por Mitógenos , Proteína de Unión al GTP rhoA/genética
8.
J Bone Miner Res ; 16(5): 832-45, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11341328

RESUMEN

Apert (Ap) syndrome is characterized by premature cranial suture ossification caused by fibroblast growth factor receptor 2 (FGFR-2) mutations. We studied the role of cadherins and signaling events in the phenotypic alterations induced by the Ap FGFR-2 S252W mutation in mutant immortalized fetal human calvaria osteoblasts. The FGFR-2 mutation caused increased expression of the osteoblast markers alkaline phosphatase (ALP), type 1 collagen (COLIA1), and osteocalcin (OC) in long-term culture. The mutation also increased cell-cell aggregation, which was suppressed by specific neutralizing anti-N- and anti-E-cadherin antibodies. Mutant osteoblasts showed increased N- and E-cadherin, but not N-cell adhesion molecule (N-CAM) messenger RNA (mRNA) and protein levels. This was confirmed in vivo by the abundant immunoreactive N- and E-cadherins in preosteoblasts in the Ap suture whereas N-CAM and alpha- and beta-catenins were unaffected. Neutralizing anti-N-cadherin antibody or N-cadherin antisense (AS) oligonucleotides but not anti-E-cadherin antibody or AS reduced ALP activity as well as ALP, COLIA1, and OC mRNA overexpression in mutant osteoblasts. Analysis of signal transduction revealed increased phospholipase Cgamma (PLCgamma) and protein kinase Calpha (PKCalpha) phosphorylation and increased PKC activity in mutant cells in basal conditions. Inhibition of PKC by calphostin C or the PKCalpha-specific inhibitor Gö6976 suppressed the increased N-cadherin mRNA and protein levels as well as the overexpression of ALP, COLIA1, and OC mRNA in mutant cells. Thus, N-cadherin plays a role in the activation of osteoblast differentiation marker genes in mutant osteoblasts and PKCalpha signaling appears to be involved in the increased N-cadherin and osteoblast gene expression induced by the S252W FGFR-2 mutation in human osteoblasts.


Asunto(s)
Acrocefalosindactilia/genética , Fosfatasa Alcalina/genética , Cadherinas/fisiología , Colágeno/genética , Regulación de la Expresión Génica , Isoenzimas/fisiología , Osteoblastos/metabolismo , Osteocalcina/genética , Proteína Quinasa C/fisiología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Acrocefalosindactilia/metabolismo , Biomarcadores , Cadherinas/genética , Cadherinas/metabolismo , Agregación Celular , Humanos , Isoenzimas/metabolismo , Mutagénesis Sitio-Dirigida , Fosforilación , Proteína Quinasa C/metabolismo , Proteína Quinasa C-alfa , Proteínas Tirosina Quinasas Receptoras/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/genética , Cráneo/citología , Activación Transcripcional
9.
J Bone Miner Res ; 18(11): 2060-8, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14606520

RESUMEN

UNLABELLED: Signaling involved in osteoblastic cell differentiation remains largely unknown. This study further investigates mechanisms involved in BMP-2-induced osteoblastic cell differentiation. We report that BMP-2 can activate JNK and p38 in osteoblastic cells and provide evidences that these MAP kinases have distinct roles in regulating alkaline phosphatase and osteocalcin expression. INTRODUCTION: Bone morphogenetic protein (BMP)-2 exerts many of its biological effects through activation of the Smad pathway. Cooperative interactions between the Smads and the stress-activated protein kinase (SAPK) p38 and c-Jun-NH2-terminal kinase (JNK) pathways have recently been observed in TGF-beta signaling. MATERIALS AND METHODS: Activation of mitogen-activated protein (MAP) kinases by BMP-2 and the role of these signaling pathways for cell differentiation induced by BMP-2 was investigated in mouse MC3T3-E1 and primary cultured calvaria-derived osteoblastic cells using immunoprecipitation, in vitro kinase assay and Western blot analysis, as well as specific MAP kinase inhibitors. RESULTS: Associated with the rapid activation of Smads, BMP-2 barely affected extracellular-signal regulated kinase (ERK) activity, whereas it induced a transient activation of p38 and JNK. The role of p38 and JNK in mediating BMP-2-induced stimulation of osteoblastic cell differentiation was evaluated using the respective specific inhibitors SB203580 and SP600125. Inhibition of p38 by SB203580 was mainly associated with decreased alkaline phosphatase (ALP) activity, whereas inhibition of JNK by SP600125 was associated with a marked reduction in osteocalcin (OC) production induced by BMP-2. Corresponding alterations in ALP and OC mRNA levels were found in cells treated with BMP-2 and inhibitors, suggesting an implication of p38 and JNK pathways in BMP-2-induced osteoblastic cell differentiation at a transcriptional level. CONCLUSION: Data presented in this study describe p38 and JNK as new signaling pathways involved in BMP-2-induced osteoblastic cell differentiation with evidences for a distinct role of each MAP kinase in the control of alkaline phosphatase and osteocalcin expression.


Asunto(s)
Proteínas Morfogenéticas Óseas/farmacología , Diferenciación Celular/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Factor de Crecimiento Transformador beta , Células 3T3 , Animales , Antracenos/farmacología , Proteína Morfogenética Ósea 2 , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos , Ratones , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Osteoblastos/enzimología , Osteoblastos/metabolismo , Cráneo/citología , Proteínas Quinasas p38 Activadas por Mitógenos
10.
Endocrinology ; 132(1): 205-11, 1993 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-7678214

RESUMEN

The equine (e) placental glycoprotein hormone eCG plays a critical though not completely understood role during the first trimester of gestation in mares. In the present work, we have developed immunoradiometric assays (m-IRMAs) for detection of eCG, eCG alpha, and eCG beta using combinations of monoclonal antibodies (mAbs) specific for epitopes that reside on free and/or combined subunits. The free eCG alpha m-IRMA was based on AHT20 mAb, specific for the free alpha-subunit of all species, and 125I-labeled ECG01 mAb, which recognizes both free and combined alpha-subunit from equine and primate species. The free eCG beta was measured by the combination of FBT11 mAb, which binds to free beta-subunit from human and equine species, and radiolabeled 518B7 mAb, which detects CG/LH from diverse mammalian species. This assay provided a better sensitivity for eLH beta than for eCG beta. However, after treatment with neuraminidase, the latter molecule was recognized as well as eLH beta, indicating that the carbohydrate structure influenced the binding of mAbs. Detection of eCG was based on the combination of ECG01 mAb (anti-alpha) as capture antibody and 125I-labeled 518B7 mAb (anti-beta). Using such assays, we measured plasma and urinary concentrations of both eCG and free subunits in pregnant mares from mating to day 90 of gestation. eCG was constantly detectable in the serum between days 40-90, as previously reported, but small amounts of the dimeric hormone in the urine were also detectable. Further, m-IRMA specific for the free beta-subunit showed that low levels (5-100 ng/ml) of eCG beta may coexist with eCG in serum and urine during early pregnancy in mares. In contrast, free eCG alpha subunit was undetectable during the first 10 weeks of gestation. These results suggested that eCG and free subunit production in pregnant mares at the beginning of gestation is similar to that observed in pregnant women. These immunoassays, specific for either intact hormone or its free subunits, constitute useful diagnostic tools for investigating reproductive problems in mares.


Asunto(s)
Gonadotropina Coriónica/metabolismo , Gonadotropinas Equinas/metabolismo , Caballos/metabolismo , Preñez/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos , Carbohidratos/inmunología , Gonadotropina Coriónica/sangre , Gonadotropina Coriónica/orina , Epítopos/inmunología , Femenino , Gonadotropinas Equinas/sangre , Gonadotropinas Equinas/orina , Ensayo Inmunorradiométrico , Hormona Luteinizante/inmunología , Sustancias Macromoleculares , Embarazo , Control de Calidad , Radioinmunoensayo
11.
Arch Mal Coeur Vaiss ; 73(10): 1145-53, 1980 Oct.
Artículo en Francés | MEDLINE | ID: mdl-6778406

RESUMEN

The authors review the literature on arrhythmias and ST changes, the principal causes of sudden death in the mid systolic click-late systolic murmur (SC-SM) syndrome, having recorded the ECG during sudden death of a patient with this syndrome by Holter monitoring. This would also appear to be the first reported case of sudden death recorded by this technique. It showed that death was caused by ventricular fibrillation which was preceded by a clinical and electrical episode of Prinzmetal angina. Hypotheses explaining the abnormalities of ventricular repolarisation and rhythm in so-called "idiopathic" SC-SM syndrome are reviewed. Special attention is paid to studies suggesting the participation of coronary spasm and autonomic nervous system disturbances. In conclusion, the authors underline: - the value of ambulatory electrocardiography in patients with the SC-SM syndrome and especially in a defined high risk group; - the value of testing for coronary spasm when coronary angiography is envisaged; - the need for better management with antiarrhythmic therapy. The indications of betablockade are discussed as these drugs are thought to favourise coronary spasm.


Asunto(s)
Angina Pectoris Variable/complicaciones , Angina de Pecho/complicaciones , Muerte Súbita/etiología , Prolapso de la Válvula Mitral/complicaciones , Angina Pectoris Variable/fisiopatología , Arritmias Cardíacas/complicaciones , Electrocardiografía , Ventrículos Cardíacos , Humanos , Masculino , Métodos , Persona de Mediana Edad , Prolapso de la Válvula Mitral/fisiopatología
12.
Rev Med Interne ; 18(10): 765-8, 1997.
Artículo en Francés | MEDLINE | ID: mdl-9500009

RESUMEN

The blinded study of CBUE (cytobacteriological urine examination) test strips was carried out in 100 patients (65 women, 35 men), mean age 85.52 +/- 6.28 years, admitted for a short stay in a geriatrics department. Urinary infection was present in 19% of the cases. Typical clinical signs (pollakiuria, dysuria, hematuria, burning upon voiding) were rare, less than 10%. General signs, such as deterioration of general status (65% of the cases) and loss of autonomy (53% of the cases), on the other hand, were predominant and their relatively unspecific nature required a reliable screening test for urinary infection. Good results of sensitivity (89.5%), specificity (98.7%), and the leukocytes/nitrites values, confirmed the validity of the urine test strip in hospitalized elderly patients. In addition, the negative predictive value of the combination leukocytes and nitrites (90.8%) would have prevented 71 samples from being sent to the laboratory for CBUE, resulting in an obvious financial savings.


Asunto(s)
Geriatría , Tiras Reactivas , Infecciones Urinarias/orina , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Infecciones Urinarias/diagnóstico
13.
J Biol Chem ; 276(31): 29028-36, 2001 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-11395480

RESUMEN

Bone morphogenetic protein-2 (BMP-2), a member of the transforming growth factor-beta (TGF-beta) family, regulates osteoblast differentiation and bone formation. Here we show a novel function of BMP-2 in human osteoblasts and identify a signaling pathway involved in this function. BMP-2 promotes apoptosis in primary human calvaria osteoblasts and in immortalized human neonatal calvaria osteoblasts, as shown by terminal deoxynucleotidyl transferase-mediated nick end labeling analysis. In contrast, TGF-beta 2 inhibits apoptosis in human osteoblasts. Studies of the mechanisms of action showed that BMP-2 increases the Bax/Bcl-2 ratio, whereas TG beta-2 has a negative effect. Moreover, BMP-2 increases the release of mitochondrial cytochrome c to the cytosol. Consistent with these results, BMP-2 increases caspase-9 and caspase-3, -6, and -7 activity, and an anti-caspase-9 agent suppresses BMP-2-induced apoptosis. Overexpression of dominant-negative Smad1 effectively blocks BMP-2-induced expression of the osteoblast transcription factor Runx2 but not the activation of caspases or apoptosis induced by BMP-2, indicating that the Smad1 signaling pathway is not involved in the BMP-2-induced apoptosis. The proapoptotic effect of BMP-2 is PKC-dependent, because BMP-2 increases PKC activity, and the selective PKC inhibitor calphostin C blocks the BMP-2-induced increased Bax/Bcl-2, caspase activity, and apoptosis. In contrast, the cAMP-dependent protein kinase A inhibitor H89, the p38 MAPK inhibitor SB203580, and the MEK inhibitor PD-98059 have no effect. The results show that BMP-2 uses a Smad-independent, PKC-dependent pathway to promote apoptosis via a Bax/Bcl-2 and cytochrome c-caspase-9-caspase-3, -6, -7 cascade in human osteoblasts.


Asunto(s)
Apoptosis/fisiología , Proteínas Morfogenéticas Óseas/farmacología , Proteínas de Neoplasias , Osteoblastos/citología , Osteoblastos/fisiología , Proteína Quinasa C/metabolismo , Transducción de Señal/fisiología , Sulfonamidas , Apoptosis/efectos de los fármacos , Proteína Morfogenética Ósea 2 , Caspasas/metabolismo , Células Cultivadas , Subunidad alfa 1 del Factor de Unión al Sitio Principal , Grupo Citocromo c/metabolismo , Citosol/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Etopósido/farmacología , Flavonoides/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Etiquetado Corte-Fin in Situ , Recién Nacido , Isoquinolinas/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Naftalenos/farmacología , Osteoblastos/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Cráneo/citología , Proteínas Smad , Proteína Smad1 , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta/farmacología , Proteína X Asociada a bcl-2
14.
J Cell Biochem ; 81(1): 68-81, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11180398

RESUMEN

Fibroblast growth factors (FGFs) are important factors regulating osteogenesis. However, the early mechanisms and signaling pathways involved in FGF actions in osteoblasts are unknown. We investigated the effects of FGF-2 on cell-cell adhesion and cadherin expression and the underlying signaling pathways in immortalized human neonatal calvaria (IHNC) cells. These cells express E- and N-cadherins, as shown by immunocytochemical and Western blot analyses. rhFGF-2 increased cell-cell adhesion at 24-72 h, as measured in a cell aggregation assay, and this effect was blocked by specific neutralizing anti-N-cadherin, but not anti-E-cadherin antibodies. Accordingly, ELISA and Western blot analyses showed that rhFGF-2 (10-100 ng/ml) dose dependently increased N-cadherin but not E-cadherin protein levels. RT-PCR analysis showed that rhFGF-2 transiently increased N-cadherin mRNA levels in IHNC cells. The RNA polymerase II inhibitor 5,6-dichloro-1-beta-D-ribofuranosyl benzimidazole prevented the rhFGF-2-induced up-regulation of N-cadherin mRNA, suggesting that transcription is necessary for this effect. Analysis of signaling molecules showed evidence that PLCgamma-PKC, Src, Erk 1/2 and p38 MAPK pathways are activated by rhFGF-2 in IHNC cells. The selective PKC inhibitors calphostin C, Ro-31-8220, Gö6976 and Gö6983 abrogated the stimulatory effect of rhFGF-2 on N-cadherin mRNA levels. The src-family tyrosine kinase inhibitor PP1 also blocked rhFGF-2-promoted N-cadherin expression. In contrast, the p38 MAP kinase inhibitor SB 203580 or the MEK inhibitor PD98059 had no effect on rhFGF-2-induced N-cadherin mRNA levels. Our data indicate that FGF-2 increases N-cadherin expression and function in human calvaria osteoblasts via activation of PKC and src-kinase pathways. This study identifies N-cadherin as a previously unrecognized target gene for FGF-2 signaling pathway that regulates cell-cell adhesion in human osteoblasts.


Asunto(s)
Cadherinas/genética , Factor 2 de Crecimiento de Fibroblastos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Osteoblastos/metabolismo , Proteína Quinasa C/metabolismo , Familia-src Quinasas/metabolismo , Cadherinas/metabolismo , Línea Celular , Activación Enzimática , Humanos , Immunoblotting , Osteoblastos/enzimología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
15.
Exp Cell Res ; 256(1): 158-67, 2000 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-10739663

RESUMEN

We recently showed that the Apert Ser252Trp fibroblast growth factor receptor-2 (FGFR-2) mutation causes premature osteoblast differentiation and increased subperiosteal calvaria bone matrix formation. To gain further insight into the cellular mechanisms involved in these effects, we examined the effects of the mutation on the expression of FGFRs in relation to cell proliferation and differentiation markers in vivo and in vitro, and we analyzed the underlying signaling pathways in mutant cells. Immunohistochemical analysis of the Apert calvaria suture showed that the Ser252Trp FGFR-2 mutation increased type 1 collagen, osteocalcin, and osteopontin expression in preosteoblasts compared to normal, whereas cell growth was not affected. The premature osteoblast differentiation induced by the mutation was associated with lower than normal FGFR-2 immunolabeling, whereas FGFR-1 and FGFR-3 levels were not decreased. Immunocytochemical analysis in osteoblasts isolated from Apert coronal suture showed that the Ser252Trp mutation induced constitutive downregulation of FGFR-2 in mutant cells. Western blot analysis of FGFRs in immortalized mutant osteoblastic cells confirmed that the mutation induced FGFR-2 downregulation. FGFR-2 mRNA levels were not altered in mutant cells, indicating that FGFR-2 downregulation resulted from receptor internalization rather than from changes in receptor mRNA. The signaling pathway involved in FGFR-2 downregulation was studied using specific inhibitors of FGF signaling molecules. The selective PKC inhibitor calphostin C markedly reduced FGFR-2 protein levels in mutant cells, in contrast to the p38 MAP kinase inhibitor SB 203580 or the Erk 1,2 MAP kinase inhibitor PD-98059, showing that PKC is involved in FGFR-2 regulation, but not in FGFR-2 downregulation in mutant cells. The results indicate that the premature osteoblast differentiation induced by the FGFR-2 Ser252Trp mutation is associated with a PKC-independent downregulation of FGFR-2 in human calvaria cells.


Asunto(s)
Acrocefalosindactilia/genética , Osteoblastos/citología , Osteoblastos/fisiología , Mutación Puntual , Proteína Quinasa C/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Acrocefalosindactilia/patología , Sustitución de Aminoácidos , Diferenciación Celular , División Celular , Cartilla de ADN , Regulación hacia Abajo , Feto , Humanos , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Valores de Referencia , Serina , Cráneo/embriología , Cráneo/patología , Transcripción Genética , Triptófano
16.
J Cell Physiol ; 183(1): 117-28, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10699973

RESUMEN

Bone morphogenetic protein-2 (BMP-2) stimulates the differentiation of osteoblastic cells. However, the mechanisms involved in this effect are not well characterized. In this study, we determined the role of the cell-cell adhesion molecules N-cadherin and E-cadherin in the promotion of osteoblast differentiation by BMP-2 in immortalized human neonatal calvaria (IHNC) cells. In cells cultured in aggregates, recombinant human BMP-2 (rhBMP-2) increased messenger RNA levels for alkaline phosphatase (ALP), the osteoblast specific transcription factor Osf2/Cbfa1 and osteocalcin, and enhanced in vitro osteogenesis in long-term culture. RT-PCR, immunocytochemical, and Western blot analyses showed that IHNC cells express E-cadherin, N-cadherin, and neural cell adhesion molecule (N-CAM) mRNA and protein. Treatment with rhBMP-2 induced a rapid and transient increase in N-cadherin and E-cadherin but not N-CAM, mRNA, and protein levels. Incubation with the RNA polymerase II inhibitor 5, 6-dichloro-1-beta-D-ribofuranosyl benzimidazole prevented the upregulation of N- and E-cadherins induced by rhBMP-2, suggesting that transcription is necessary for this effect. N- and E-cadherins were functional because rhBMP-2 increased cell-cell adhesion in a cell aggregation assay, and this effect was largely blocked by N-cadherin- and E-cadherin-neutralizing antibodies. In addition, N- and E-cadherin antibodies decreased the basal ALP activity and completely suppressed the rhBMP-2-induced increase in ALP activity and mRNA levels. Furthermore, anti-N-cadherin or anti-E-cadherin antibodies markedly decreased Osf2/Cbfa1 mRNA levels and abolished the rhBMP-2-induced increased Osf2/Cbfa1 expression, and reduced the increased osteocalcin mRNA levels induced by rhBMP-2. We conclude that rhBMP-2 rapidly and transiently increases N- and E-cadherin expression, and this effect mediates the rhBMP-2-induced early promotion of cell-cell adhesion and osteoblast marker gene expression in human calvaria cells.


Asunto(s)
Proteínas Morfogenéticas Óseas/farmacología , Cadherinas/fisiología , Diferenciación Celular/fisiología , Osteoblastos/citología , Osteoblastos/fisiología , Osteogénesis/fisiología , Factor de Crecimiento Transformador beta , Proteína Morfogenética Ósea 2 , Cadherinas/genética , Adhesión Celular , Diferenciación Celular/efectos de los fármacos , División Celular , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Recién Nacido , Cinética , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Cráneo , Factores de Tiempo
17.
Exp Cell Res ; 269(1): 154-61, 2001 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-11525648

RESUMEN

Cell-cell adhesion mediated by cadherins is believed to play an essential role in the control of cell differentiation and tissue formation. Our recent studies indicate that N-cadherin is involved in human osteoblast differentiation. However, the signalling molecules that regulate cadherins in osteoblasts are not known. We tested the possibility that N-cadherin expression and function may be regulated by direct activation of protein kinase C (PKC) in human osteoblasts. Treatment of immortalized human neonatal calvaria (IHNC) cells with phorbol 12,13-dibutyrate (100 nM) transiently increased PKC activity. RT-PCR analysis showed that transient treatment with phorbol ester transiently increased N-cadherin mRNA levels at 4-12 h. Western blot analysis showed that N-cadherin protein levels were increased by phorbol ester at 24-48 h, and this was confirmed by immunocytochemical analysis. In contrast, E-cadherin expression was not affected. Transient treatment of IHNC cells with phorbol ester increased cell-cell aggregation, which was suppressed by neutralizing N-cadherin antibody, showing that the increased N-cadherin induced by phorbol ester was functional. Finally, phorbol ester dose-dependently increased alkaline phosphatase activity, an early marker of osteoblast differentiation. This effect was comparable to the promoting effect of BMP-2, a potent activator of osteoblast differentiation. These data show that direct activation of PKC by phorbol ester increases N-cadherin expression and function, and promotes ALP activity in human calvaria osteoblasts, which provides a signaling mechanism by which N-cadherin is regulated and suggests a role for PKC in N-cadherin-mediated control of human osteoblast differentiation.


Asunto(s)
Cadherinas/metabolismo , Adhesión Celular/fisiología , Comunicación Celular/fisiología , Diferenciación Celular/fisiología , Osteoblastos/enzimología , Proteína Quinasa C/metabolismo , Factor de Crecimiento Transformador beta , Regulación hacia Arriba/fisiología , Fosfatasa Alcalina/efectos de los fármacos , Fosfatasa Alcalina/metabolismo , Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas/farmacología , Cadherinas/efectos de los fármacos , Cadherinas/genética , Carcinógenos/farmacología , Adhesión Celular/efectos de los fármacos , Agregación Celular/efectos de los fármacos , Agregación Celular/fisiología , Comunicación Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular Transformada/citología , Línea Celular Transformada/efectos de los fármacos , Línea Celular Transformada/enzimología , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Humanos , Recién Nacido , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Ésteres del Forbol/farmacología , Proteína Quinasa C/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Cráneo/citología , Cráneo/efectos de los fármacos , Cráneo/enzimología , Regulación hacia Arriba/efectos de los fármacos
18.
Exp Cell Res ; 297(1): 235-46, 2004 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15194439

RESUMEN

Fibroblast growth factor-2 (FGF-2) is an important molecule that controls bone formation through activation of osteoblastic cell replication and differentiation. The role of FGF-2 on human osteoblast survival and the signaling pathway that mediates its effect are not known. We studied the effect of FGF-2 on apoptosis induced by low serum concentration and the signal transduction pathway involved in this effect in human primary calvaria osteoblasts and immortalized osteoblastic cells. Treatment with FGF-2 for 24-48 h protected against osteoblast apoptosis induced by low serum concentration, through specific inhibition of caspase-2 and caspase-3 activity. Pharmacological inhibition of MEK-1 and p38 MAPK had no effect on the inhibition of caspases-2 and -3 induced by FGF-2. In contrast, inhibition of PI3K with LY294002 abolished the FGF-2-induced inhibition of caspases-2 and -3. FGF-2 increased PI3K activity but did not induce phosphorylation of Akt or the downstream effector p70 S6 kinase. FGF-2 also induced GSK-3alpha and beta phosphorylation in osteoblastic cells, which however did not result in beta-catenin accumulation or Lef/Tcf transcriptional activity. In contrast, lithium induced beta-catenin accumulation, Lef/Tcf transcriptional activation and increased caspase-2 and -3 activity. The results indicate that the immediate protective effect of FGF-2 on human osteoblastic cell apoptosis involves PI3K and inhibition of downstream caspases, independently of GSK-3 and beta-catenin-Lef/Tcf-mediated transcription.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Osteoblastos/metabolismo , Osteogénesis/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/fisiología , Transactivadores/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Proteínas Sanguíneas/deficiencia , Caspasa 2 , Caspasa 3 , Inhibidores de Caspasas , Caspasas/metabolismo , Línea Celular Transformada , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Proteínas de Unión al ADN/agonistas , Proteínas de Unión al ADN/metabolismo , Inhibidores Enzimáticos/farmacología , Retroalimentación Fisiológica/efectos de los fármacos , Retroalimentación Fisiológica/fisiología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Glucógeno Sintasa Quinasa 3/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Litio/farmacología , Factor de Unión 1 al Potenciador Linfoide , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/agonistas , Factores de Transcripción/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , beta Catenina
19.
Am J Pathol ; 158(5): 1833-42, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11337381

RESUMEN

Apert syndrome is an autosomal dominant disorder characterized by premature cranial ossification resulting from fibroblast growth factor receptor-2 (FGFR-2)-activating mutations. We have studied the effects of the prominent S252W FGFR-2 Apert mutation on apoptosis and the underlying mechanisms in human mutant osteoblasts. In vivo analysis of terminal deoxynucleotidyl transferase-mediated nick-end labeling revealed premature apoptosis of mature osteoblasts and osteocytes in the Apert suture compared to normal coronal suture. In vitro, mutant osteoblasts showed increased apoptosis, as demonstrated by terminal deoxynucleotidyl transferase-mediated nick-end labeling analysis, trypan blue staining, and DNA fragmentation. Mutant osteoblasts also showed increased activity of caspase-8 and effector caspases (-3, -6, -7) constitutively. This was related to protein kinase C activation because the selective protein kinase C inhibitor calphostin C inhibited caspase-8, effector caspases, and apoptosis in mutant osteoblasts. Apert osteoblasts also showed increased expression of interleukin (IL)-1alpha, IL-1beta, Fas, and Bax, and decreased Bcl-2 levels. Specific neutralizing anti-IL-1 antibody reduced Fas levels, Bax expression, effector caspases activity, and apoptosis in mutant cells. Thus, the Apert S252W FGFR-2 mutation promotes apoptosis in human osteoblasts through activation of protein kinase C, overexpression of IL-1 and Fas, activation of caspase-8, and increased Bax/Bcl-2 levels, leading to increased effector caspases and DNA fragmentation. This identifies a complex FGFR-2 signaling pathway involved in the premature apoptosis induced by the Apert S252W FGFR-2 mutation in human calvaria osteoblasts.


Asunto(s)
Acrocefalosindactilia/patología , Apoptosis , Craneosinostosis/patología , Osteoblastos/patología , Acrocefalosindactilia/genética , Sustitución de Aminoácidos , Caspasa 3 , Caspasa 8 , Caspasa 9 , Caspasas/metabolismo , Células Cultivadas , Craneosinostosis/genética , Humanos , Interleucina-1/metabolismo , Interleucina-1/fisiología , Mutación , Osteoblastos/metabolismo , Proteína Quinasa C/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/genética , Proteína X Asociada a bcl-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA