Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oral Dis ; 24(6): 1037-1041, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29566451

RESUMEN

OBJECTIVE: To identify the molecular genetic etiology of an individual with a dysmorphic face, unusual teeth mobility, and root resorption. SUBJECTS AND METHODS: DNA samples were collected from a trio of family members, and whole-exome sequencing was performed. RESULTS: Mutational analysis revealed a de novo mutation (c.6787C>T) in the last exon of the NOTCH2 gene. This mutation would introduce a premature stop codon [p.(Gln2263*)] and generate a truncated protein without C-terminus, escaping from the nonsense-mediated decay system. Sanger sequencing confirmed that this mutation was generated spontaneously. CONCLUSIONS: In this study, we identified a novel nonsense mutation in the last exon of the NOTCH2 gene causing Hajdu-Cheney syndrome. We described the genotype and phenotype correlation and the related dental complications. These results will advance the understanding of the NOTCH2 signaling in periodontitis and root resorption.


Asunto(s)
Síndrome de Hajdu-Cheney/genética , Receptor Notch2/genética , Adolescente , Niño , Preescolar , Codón sin Sentido , Femenino , Síndrome de Hajdu-Cheney/complicaciones , Síndrome de Hajdu-Cheney/diagnóstico , Humanos , Maloclusión/etiología , Resorción Radicular/etiología , Movilidad Dentaria/etiología
2.
J Dent Res ; 97(9): 1064-1069, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29554435

RESUMEN

Tooth enamel, the hardest tissue in the human body, is formed after a complex series of interactions between dental epithelial tissue and the underlying ectomesenchyme. Nonsyndromic amelogenesis imperfecta (AI) is a rare genetic disorder affecting tooth enamel without other nonoral symptoms. In this study, we identified 2 novel ENAM mutations in 2 families with hypoplastic AI by whole exome sequencing. Family 1 had a heterozygous splicing donor site mutation in intron 4, NM_031889; c.123+2T>G. Affected individuals had hypoplastic enamel with or without the characteristic horizontal hypoplastic grooves in some teeth. Family 2 had a nonsense mutation in the last exon, c.1842C>G, p.(Tyr614*), that was predicted to truncate the protein by 500 amino acids. Participating individuals had at least 1 mutant allele, while the proband had a homozygous mutation. Most interestingly, the clinical phenotype of the individuals harboring the heterozygous mutation varied from a lack of penetrance to a mild hypoplastic enamel defect. We believe that these findings will broaden our understanding of the clinical phenotype of AI caused by ENAM mutations.


Asunto(s)
Amelogénesis Imperfecta/genética , Proteínas de la Matriz Extracelular/genética , Mutación/genética , Niño , Consanguinidad , Femenino , Humanos , Linaje , Fenotipo , Reacción en Cadena de la Polimerasa , Turquía , Secuenciación del Exoma , Adulto Joven
3.
Oral Dis ; 22(4): 297-302, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26762616

RESUMEN

OBJECTIVE: To identify the molecular genetic aetiology of a family with autosomal dominant amelogenesis imperfecta (AI). SUBJECTS AND METHODS: DNA samples were collected from a six-generation family, and the candidate gene approach was used to screen for the enamelin (ENAM) gene. Whole-exome sequencing and linkage analysis with SNP array data identified linked regions, and candidate gene screening was performed. RESULTS: Mutational analysis revealed a mutation (c.561_562delCT and p.Tyr188Glnfs*13) in the DLX3 gene. After finding a recurrent DLX3 mutation, the clinical phenotype of the family members was re-examined. The proband's mother had pulp elongation in the third molars. The proband had not hair phenotype, but her cousin had curly hair at birth. CONCLUSIONS: In this study, we identified a recurrent 2-bp deletional DLX3 mutation in a new family. The clinical phenotype was the mildest one associated with the DLX3 mutations. These results will advance the understanding of the functional role of DLX3 in developmental processes.


Asunto(s)
Amelogénesis Imperfecta/genética , Secuencia de Bases , Proteínas de Homeodominio/genética , Eliminación de Secuencia , Factores de Transcripción/genética , Adulto , Amelogénesis Imperfecta/diagnóstico por imagen , Niño , Análisis Mutacional de ADN , Exoma , Femenino , Humanos , Masculino , Linaje , Fenotipo , Radiografía Dental
4.
J Dent Res ; 94(8): 1063-9, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26124219

RESUMEN

In order to achieve highly mineralized tooth enamel, enamel proteinases serve the important function of removing the remaining organic matrix in the mineralization and maturation of the enamel matrix. Mutations in the kallikrein 4 (KLK4), enamelysin (MMP20), and WDR72 genes have been identified as causing hypomaturation enamel defects in an autosomal-recessive hereditary pattern. In this report, 2 consanguineous families with a hypomaturation-type enamel defect were recruited, and mutational analysis was performed to determine the molecular genetic etiology of the disease. Whole exome sequencing and autozygosity mapping identified novel homozygous mutations in the KLK4 (c.620_621delCT, p.Ser207Trpfs*38) and MMP20 (c.1054G>A, p.Glu352Lys) genes. Further analysis on the effect of the mutations on the translation, secretion, and function of KLK4 and MMP20 revealed that mutant KLK4 was degraded intracellularly and became inactive while mutant MMP20 was expressed at a normal level but secreted only minimally with proteolytic function.


Asunto(s)
Amelogénesis Imperfecta/genética , Calicreínas/genética , Metaloproteinasa 20 de la Matriz/genética , Mutación , Amelogénesis Imperfecta/diagnóstico por imagen , Western Blotting , Niño , Consanguinidad , Femenino , Genotipo , Homocigoto , Humanos , Linaje , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple , Radiografía Panorámica , Análisis de Secuencia de ADN
5.
Oral Dis ; 21(4): 456-61, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25431241

RESUMEN

OBJECTIVE: Hereditary defects in tooth enamel formation, amelogenesis imperfecta (AI), can be non-syndromic or syndromic phenotype. Integrins are signaling proteins that mediate cell-cell and cell-extracellular matrix communication, and their involvement in tooth development is well known. The purposes of this study were to identify genetic cause of an AI family and molecular pathogenesis underlying defective enamel formation. MATERIALS AND METHODS: We recruited a Turkish family with isolated AI and performed mutational analyses to clarify the underlying molecular genetic etiology. RESULTS: Autozygosity mapping and exome sequencing identified a novel homozygous ITGB6 transversion mutation in exon 4 (c.517G>C, p.Gly173Arg). The glycine at this position in the middle of the ßI-domain is conserved among a wide range of vertebrate orthologs and human paralogs. Clinically, the enamel was generally thin and pitted with pigmentation. Thicker enamel was noted at the cervical area of the molars. CONCLUSIONS: In this study, we identified a novel homozygous ITGB6 mutation causing isolated AI, and this advances the understanding of normal and pathologic enamel development.


Asunto(s)
Amelogénesis Imperfecta/etiología , Amelogénesis Imperfecta/genética , Cadenas beta de Integrinas/genética , Mutación , Amelogénesis/genética , Amelogénesis Imperfecta/sangre , Amelogénesis Imperfecta/diagnóstico por imagen , Niño , Análisis Mutacional de ADN , Exones , Femenino , Humanos , Diente Molar/patología , Linaje , Reacción en Cadena de la Polimerasa , Alineación de Secuencia , Turquía
6.
J Dent Res ; 93(10): 988-92, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25143514

RESUMEN

Amelogenesis imperfecta (AI) is a genetic disease affecting tooth enamel formation. AI can be an isolated entity or a phenotype of syndromes. To date, more than 10 genes have been associated with various forms of AI. We have identified 2 unrelated Turkish families with hypoplastic AI and performed mutational analysis. Whole-exome sequencing identified 2 novel heterozygous nonsense mutations in the ENAM gene (c.454G>T p.Glu152* in family 1, c.358C>T p.Gln120* in family 2) in the probands. Affected individuals were heterozygous for the mutation in each family. Segregation analysis within each family revealed individuals with incomplete penetrance or extremely mild enamel phenotype, in spite of having the same mutation with the other affected individuals. We believe that these findings will broaden our understanding of the clinical phenotype of AI caused by ENAM mutations.


Asunto(s)
Amelogénesis Imperfecta/genética , Codón sin Sentido/genética , Proteínas de la Matriz Extracelular/genética , Penetrancia , Adolescente , Niño , Segregación Cromosómica/genética , Codón de Terminación/genética , Consanguinidad , Citosina , Esmalte Dental/patología , Hipoplasia del Esmalte Dental/genética , Exoma/genética , Exones/genética , Femenino , Ácido Glutámico/genética , Glutamina/genética , Guanina , Heterocigoto , Humanos , Masculino , Fenotipo , Timina
7.
J Dent Res ; 93(10): 980-7, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25117480

RESUMEN

Tooth enamel is the most highly mineralized tissue in vertebrates. Enamel crystal formation and elongation should be well controlled to achieve an exceptional hardness and a compact microstructure. Enamel matrix calcification occurs with several matrix proteins, such as amelogenin, enamelin, and ameloblastin. Among them, amelogenin is the most abundant enamel matrix protein, and multiple isoforms resulting from extensive but well-conserved alternative splicing and postsecretional processing have been identified. In this report, we recruited a family with a unique enamel defect and identified a silent mutation in exon 4 of the AMELX gene. We show that the mutation caused the inclusion of exon 4, which is almost always skipped, in the mRNA transcript. We further show, by generating and characterizing a transgenic animal model, that the alteration of the ratio and quantity of the developmentally conserved alternative splicing repertoire of AMELX caused defects in enamel matrix mineralization.


Asunto(s)
Empalme Alternativo/genética , Amelogénesis Imperfecta/genética , Amelogenina/genética , Secuencia Conservada/genética , ARN Mensajero/genética , Ameloblastos/patología , Amelogénesis Imperfecta/patología , Animales , Niño , Cristalografía , Esmalte Dental/patología , Esmalte Dental/ultraestructura , Exones/genética , Femenino , Ligamiento Genético , Vectores Genéticos/genética , Humanos , Intrones/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Transgénicos , Repeticiones de Microsatélite/genética , Mutación/genética , Fenotipo , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas/genética , Transcripción Genética/genética
8.
J Dent Res ; 93(4): 366-70, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24532815

RESUMEN

Amelogenesis imperfecta is a heterogeneous group of genetic conditions affecting enamel formation. Recently, mutations in solute carrier family 24 member 4 (SLC24A4) have been identified to cause autosomal recessive hypomaturation amelogenesis imperfecta. We recruited a consanguineous family with hypomaturation amelogenesis imperfecta with generalized brown discoloration. Sequencing of the candidate genes identified a 10-kb deletion, including exons 15, 16, and most of the last exon of the SLC24A4 gene. Interestingly, this deletion was caused by homologous recombination between two 354-bp-long homologous sequences located in intron 14 and the 3' UTR. This is the first report of exonal deletion in SLC24A4 providing confirmatory evidence that the function of SLC24A4 in calcium transport has a crucial role in the maturation stage of amelogenesis.


Asunto(s)
Amelogénesis Imperfecta/genética , Antiportadores/genética , Exones/genética , Eliminación de Gen , Regiones no Traducidas 3'/genética , Emparejamiento Base/genética , Niño , Hibridación Genómica Comparativa , Consanguinidad , Femenino , Homocigoto , Humanos , Intrones/genética , Linaje
9.
Gene Ther ; 17(8): 1052-61, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20485381

RESUMEN

Cell-permeable peptides (CPPs) promote the transduction of nonpermissive cells by recombinant adenovirus (rAd) to improve the therapeutic efficacy of rAd. In this study, branched oligomerization of CPPs significantly enhanced the transduction of human mesenchymal stem cells (MSCs) by rAd in a CPP type-independent manner. In particular, tetrameric CPPs increased transduction efficiency at 3000-5000-fold lower concentrations than did monomeric CPPs. Although branched oligomerization of CPPs also increases cytotoxicity, optimal concentrations of tetrameric CPPs required for maximum transduction are at least 300-1000-fold lower than those causing 50% cytotoxicity. Furthermore, although only approximately 60% of MSCs were maximally transduced at 500 muM of monomeric CPPs, >95% of MSCs were transduced with 0.1 muM of tetrameric CPPs. Tetrameric CPPs also significantly increased the formation and net surface charge of CPP/rAd complexes, as well as the binding of rAd to cell membranes at a greater degree than did monomeric CPPs, followed by rapid internalization into MSCs. In a critical-size calvarial defect model, the inclusion of tetrameric CPPs in ex vivo transduction of rAd expressing bone morphogenetic protein 2 into MSCs promoted highly mineralized bone formation. In addition, MSCs that were transduced with rAd expressing brain-derived neurotrophic factor in the presence of tetrameric CPPs improved functional recovery in a spinal cord injury model. These results demonstrated the potential for tetrameric CPPs to provide an innovative tool for MSC-based gene therapy and for in vitro gene delivery to MSCs.


Asunto(s)
Adenoviridae/genética , Péptidos de Penetración Celular/química , Terapia Genética/métodos , Células Madre Mesenquimatosas/metabolismo , Transducción Genética/métodos , Animales , Enfermedades Óseas/genética , Enfermedades Óseas/terapia , Proteína Morfogenética Ósea 2/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Osteogénesis/genética , Ratas , Ratas Sprague-Dawley , Cráneo/crecimiento & desarrollo
10.
Cell Death Differ ; 16(10): 1332-43, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19543237

RESUMEN

Interleukin-17 (IL-17) is a cytokine secreted primarily by T(H)-17 cells. Although IL-17 is primarily associated with the induction of tissue inflammation, the other biological roles of IL-17, including non-immune functions, have yet to be thoroughly explored. Here, we report that T-cell-produced IL-17 can induce proliferation of human bone marrow-derived mesenchymal stem cells (hMSCs) in a manner dependent on the generation of reactive oxygen species (ROS). Rac1 GTPase and NADPH oxidase 1 (Nox1) are activated by IL-17 to produce ROS, which in turn stimulates hMSC proliferation. The activation of the MEK-ERK pathway is also crucial for IL-17-dependent hMSC proliferation. TRAF6 and Act1 are required to activate Nox 1 and to phosphorylate MEK on IL-17 stimulation. Interestingly, IL-17 not only accelerates the proliferation of hMSCs, but also induces their migration, motility, and osteoblastic differentiation. Furthermore, IL-17 induces the expression of M-CSF and receptor activator of NF-kappaB ligand (RANKL) on hMSCs, thereby supporting osteoclastogenesis both in vivo and in vitro. On the basis of these results, we suggest that IL-17 can function as a signal to induce extensive bone turnover by regulating hMSC recruitment, proliferation, motility, and differentiation.


Asunto(s)
Remodelación Ósea , Diferenciación Celular , Proliferación Celular , Interleucina-17/metabolismo , Células Madre Mesenquimatosas/citología , Azacitidina/farmacología , Movimiento Celular , Conexina 43/genética , Conexina 43/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Ciclooxigenasa 2/metabolismo , Humanos , Factor Estimulante de Colonias de Macrófagos/metabolismo , Células Madre Mesenquimatosas/metabolismo , NADPH Oxidasa 1 , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Ligando RANK/metabolismo , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo
11.
J Dent Res ; 87(4): 354-7, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18362318

RESUMEN

Tricho-dento-osseous syndrome (TDO) is an autosomal-dominant disease characterized by curly hair at birth, enamel hypoplasia, taurodontism, and a thick cortical bone. A common DLX3 gene mutation (c.571_574delGGGG) has been identified in multiple families with variable clinical phenotypes. Recently, another DLX3 gene mutation (c.561_562delCT) was reported to cause amelogenesis imperfecta with taurodontism (AIHHT). We identified a Korean family with overlapping phenotypes of TDO and AIHHT. We performed mutational analysis to discover its genetic etiology. The identified mutation was c.561_562delCT mutation in the DLX3 gene. The enamel was hypomature and hypoplastic. The characteristic taurodontic features were not identified. Increased bone density or thickness could not be revealed by cephalometric, hand-wrist, and panoramic radiographs. Affected individuals reported that their nails were brittle, and they had curly hair at birth. This study clearly showed that the c.561_562delCT mutation had not only enamel defects, but also other clinical phenotypes resembling those of TDO syndrome.


Asunto(s)
Mutación del Sistema de Lectura/genética , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Amelogénesis Imperfecta/genética , Niño , Codón/genética , Cisteína/genética , Hipoplasia del Esmalte Dental/genética , Cavidad Pulpar/anomalías , Cabello/anomalías , Humanos , Masculino , Uñas Malformadas/genética , Linaje , Fenotipo , Eliminación de Secuencia/genética , Síndrome , Treonina/genética
12.
Cell Death Differ ; 13(11): 1879-91, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16498455

RESUMEN

Osteoclast (Oc) differentiation is fundamentally controlled by receptor activator of nuclear factor kappaB ligand (RANKL). RANKL signalling targets include mitogen-activated protein kinases (MAPKs), nuclear factor kappaB (NF-kappaB), and nuclear factor of activated T cells (NFAT)c1. In this study, we found that p38 MAPK upstream components transforming growth factor-beta-activated kinase 1 (TAK1), MKK3, and MKK6 increased by RANKL in an early stage of osteoclastogenesis from primary bone marrow cells, which led to enhanced p38 activation. Retroviral transduction of dominant-negative (DN) forms of TAK1 and MKK6, but not that of MKK3, reduced Oc differentiation. Transduction of TAK1-DN and MKK6-DN and treatment with the p38 inhibitor SB203580 attenuated NFATc1 induction by RANKL. TAK1-DN, MKK6-DN, and SB203580, but not MKK3-DN, also suppressed RANKL stimulation of NF-kappaB transcription activity in a manner dependent on p65 phosphorylation on Ser-536. These results indicate that TAK1 and MKK6 constitute the p38 signalling pathway to participate to Oc differentiation by RANKL through p65 phosphorylation and NFATc1 induction, and that MKK6 and MKK3 have differential roles in osteoclastogenesis from bone marrow precursors.


Asunto(s)
Diferenciación Celular/fisiología , MAP Quinasa Quinasa 6/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , FN-kappa B/metabolismo , Osteoclastos/citología , Ligando RANK/farmacología , Activación Transcripcional/efectos de los fármacos , Animales , Células Cultivadas , Activación Enzimática/efectos de los fármacos , MAP Quinasa Quinasa 3/antagonistas & inhibidores , MAP Quinasa Quinasa 6/antagonistas & inhibidores , Ratones , Factores de Transcripción NFATC/metabolismo , Osteogénesis/efectos de los fármacos , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Transducción de Señal/efectos de los fármacos , Células Madre/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
Bone ; 32(3): 217-27, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12667549

RESUMEN

Protein kinase C (PKC) proteins have been shown to be involved in diverse cellular responses of various cell types. In experiments to identify genes regulated during osteoclast differentiation by a cDNA microarray approach, we found that the gene expression of PKC-betaII was upregulated in differentiated cells. Reverse transcription-polymerase chain reaction and Western blotting analyses also showed an increase in PKC-betaI as well as PKC-betaII during osteoclast formation in mouse bone marrow cell cultures in the presence of macrophage-colony stimulating factor (M-CSF) and receptor activator of nuclear factor-kappaB ligand (RANKL). Use of an antisense oligonucleotide to PKC-betaII resulted in a reduction in the RANKL-driven osteoclastogenesis. Pharmacological intervention with PKC-beta activity by the specific inhibitor CG53353 suppressed cellular differentiation and fusion processes during osteoclastogenesis and inhibited bone-resorbing function of mature osteoclasts. PKC-beta inhibition abolished the ERK and MEK activation by macrophage-colony stimulating factor and RANKL in osteoclast precursor cells whereas the cytokine-induced NF-kappaB activation was not hampered by the PKC-beta inhibition. Our findings indicate that PKC-beta has a role in regulation of osteoclast formation and function potentially by participating in the ERK signaling pathway of M-CSF and RANKL.


Asunto(s)
Osteoclastos/citología , Osteoclastos/enzimología , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Animales , Resorción Ósea/metabolismo , Resorción Ósea/fisiopatología , Proteínas Portadoras/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Factor Estimulante de Colonias de Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos ICR , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C beta , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B
14.
Immunopharmacol Immunotoxicol ; 24(1): 31-41, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12022443

RESUMEN

Expression of alkaline phosphatase(ALP)activity represents a key event during the differentiation processes of osteoblasts, and the level of ALP activity has been routinely used as a relative measure of differentiation stages of osteoblasts. In human osteoblasts, we showed that vitamin D3 analogue, 1,25(OH)2D3, had a stimulatory effect on ALP activity after 3 days, compared with control. The treatment of PD098059, an ERK MAP Kinase inhibitor, had a reducing effect on ALP activity, a differentiation marker in 1,25(OH)2D3-treated primary human osteoblasts. However, SB203580, a potent p38 MAP Kinase inhibitor, had no effect on the differentiation in this system. This indicates that ERK, not p38, is directly related to 1,25(OH)2D3-stimulated ALP activity in primary human osteoblasts. These results also show that the vitamin D3 analogue stimulates ERK1 activation in primary human osteoblasts. This finding provides one of signaling pathways for differentiation in primary human osteoblasts.


Asunto(s)
Calcitriol/farmacología , Agonistas de los Canales de Calcio/farmacología , Diferenciación Celular/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Osteoblastos/enzimología , Fosfatasa Alcalina/metabolismo , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Humanos , Imidazoles/farmacología , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/inmunología , Osteoblastos/citología , Pruebas de Precipitina , Piridinas/farmacología
15.
Bone ; 30(1): 71-7, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11792567

RESUMEN

Phosphatidylinositol 3-kinase (PI 3-kinase) and mitogen-activated protein kinases (MAPKs) have been implicated in diverse cellular functions, including proliferation, migration, and survival. In this study, we examined the involvement of these kinases in osteoclast differentiation by employing specific inhibitors of the kinases. The osteoclast differentiation was assessed in three different culture systems: a coculture of mouse bone marrow cells with mouse calvarial osteoblasts, a mouse bone marrow cell culture in the presence of receptor activator of NF-kappaB ligand (RANKL) and macrophage-colony stimulating factor (M-CSF), and a culture of bone-resident osteoclast precursor cells driven by RANKL and M-CSF. LY294002, a specific inhibitor of PI 3-kinase, potently inhibited osteoclast differentiation in all culture systems when assessed by both tartrate-resistant acid phosphatase (TRAP) staining and dentine resorption assays. Inhibition of p38 MAPK by SB202190 resulted in a strong suppression in the exogenous RANKL dependent mouse bone marrow and bone resident precursor cell cultures. Another MAPK pathway inhibitor (PD98059), which blocks the activation of extracellular signal-regulated kinase (ERK) by inhibiting the upstream kinase MAPK-ERK kinase (MEK) 1, exerted an inhibitory effect on osteoclast differentiation only at the highest concentration tested (30 micromol/L) in many cases. Whether the signaling pathways involving these kinases are activated by RANKL was also examined. The RANKL-stimulated phosphorylation of Akt, a downstream target of PI 3-kinase, and that of ERK were observed. RANKL also stimulated the activity of p38. These results suggest that PI 3 kinase, p38, and ERK play roles in osteoclast differentiation, at least in part, by participating in RANKL signaling.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/metabolismo , Osteoclastos/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Proteínas Portadoras/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Cromonas/farmacología , Técnicas de Cocultivo , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Humanos , Imidazoles/farmacología , MAP Quinasa Quinasa 1 , Factor Estimulante de Colonias de Macrófagos/farmacología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos ICR , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Morfolinas/farmacología , Osteoblastos/citología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Piridinas/farmacología , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Transducción de Señal , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
16.
J Biol Chem ; 276(52): 49343-9, 2001 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-11675379

RESUMEN

Differentiated osteoclasts have a short life span. We tested various cytokines and growth factors for the effects on the survival of purified mature osteoclasts. In the absence of any added factors, osteoclasts exhibited the survival rate of less than 25% after a 24-h incubation. Among the tested factors, tumor necrosis factor-alpha (TNF-alpha) was found to increase the survival rate to approximately 80%. The TNF-alpha-enhanced survival of osteoclasts appeared to be associated with reduction in apoptosis and suppression of caspase activation. The antiapoptotic signaling pathways involved in the TNF-alpha-induced osteoclast survival were investigated. TNF-alpha treatment increased the phosphorylation of Akt in osteoclasts, which was suppressed by a phosphatidylinositol 3-kinase inhibitor LY294002 and an Src family kinase-selective inhibitor PP1. These inhibitors also attenuated the TNF-alpha stimulation of osteoclast survival. In addition an increase in the phosphorylation of ERK was observed upon TNF-alpha stimulation. PD98059, a specific inhibitor of the ERK-activating kinase MEK-1, abolished the TNF-alpha-induced ERK phosphorylation and osteoclast survival, and in these responses the involvement of Grb2 and ceramide was observed. These results suggest that TNF-alpha promotes the survival of osteoclasts by engaging the phosphatidylinositol 3-kinase Akt and MEK/ERK signaling pathways.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Osteoclastos/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Animales , Apoptosis/efectos de los fármacos , Proteínas Portadoras/farmacología , Caspasas/metabolismo , Supervivencia Celular , Células Cultivadas , Ceramidas/metabolismo , Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Proteína Adaptadora GRB2 , Humanos , Interleucina-1/farmacología , MAP Quinasa Quinasa 1 , Glicoproteínas de Membrana/farmacología , Morfolinas/farmacología , Osteoclastos/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas/metabolismo , Proteínas/farmacología , Proteínas Proto-Oncogénicas c-akt , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Transducción de Señal/fisiología
17.
Immunopharmacol Immunotoxicol ; 23(2): 133-52, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11417843

RESUMEN

The aim of this study is to elucidate the possible mechanism of apoptosis in response to hypoxia in MC3T3E1 osteoblasts. MC3T3E1 osteoblasts under hypoxic conditions (2% oxygen) resulted in apoptosis in a time-dependent manner estimated by DNA fragmentation assay and nuclear morphologystained with fluorescent dye, Hoechst 33258. Pretreatment with Z-VAD-FMK, a pan-caspase inhibitor, or Z-DEVD-CHO, a specific caspase-3 inhibitor, completely suppressed the DNA ladder in response to hypoxia. An increase in caspase-3-like protease (DEVDase) activity was observed during apoptosis, but no caspase- activity (YVADase) was detected. To confirm what caspases are involved in apoptosis, western blot analysis was performed using anti-caspase-3 or -6 antibody. The 10-kDa protein, corresponding to the active products of caspase-3 and the 10-kDA protein of the active protein of caspase-6 were generated in hypoxia-challenged cells in which processing of the full length form of caspase-3 and -6 was evident. With a time course similar to this caspase-3 and -6 activation was evident, hypoxic stress caused the cleavage of lamin A, typical of caspase-6 activity. In addition, the stress elicited the release of cytochrome c into the cytosol during apoptosis. Furthermore, we have observed that pre-treatment with SB203580, a selective p38 MAP kinase (p38 MAPK) inhibitor, attenuated the hypoxia-induced apoptosis. The addition of SB203580 suppressed caspase-3 and -6-like protease activity by hypoxia up to 50%. In contrast, PD98059 had no effect on the hypoxia-induced apoptosis. To confirm the involvement of MAP kinase, JNK/SAPK, ERK, or p38 kinase assay was performed. Although p38 MAPK was activated in response to hypoxic treatment, the other MAP kinase -JNK/SAPK or ERK- was not or modestly activated. These results suggest that p38 MAPK positively regulates hypoxia-induced apoptosis in MC3T3E1 osteoblasts.


Asunto(s)
Apoptosis/fisiología , Hipoxia de la Célula/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Células 3T3 , Animales , Apoptosis/efectos de los fármacos , Caspasa 3 , Caspasa 6 , Caspasas/metabolismo , Grupo Citocromo c/metabolismo , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Imidazoles/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos , Ratones , Mitocondrias/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Piridinas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos
18.
Pharmacol Res ; 43(3): 275-83, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11401420

RESUMEN

Treatment of MC3T3E-1 osteoblast cultures with combined interferon- gamma(IFN- gamma), lipopolysaccharide (LPS) and tumor necrosis factor- alpha(TNF- alpha) induces expressions of inducible nitric oxide synthase (iNOS) and interleukin-6 (IL-6), resulting in sustained releases of large amounts of nitric oxide and IL-6. However IFN- gamma, LPS and TNF- alpha individually induces non-detectable or small amounts of NO and IL-6 in MC3T3E-1 osteoblasts. The role of mitogen-activated protein kinase (MAPK) activation in the early intracellular signal transduction involved in iNOS and IL-6 transcription in the combined agents-stimulated osteoblasts has been investigated. The p38 MAPK pathway is specifically involved in the combined agents-induced NO and IL-6 release, since NO and IL-6 release in the presence of a specific inhibitor of p38 MAPK, 4-(4-fluorophenyl)-2-(4-metylsulfinylphenyl)-5-(4-pyridyl)imidazole (SB203580), are significantly diminished. In contrast, PD98059, a specific inhibitor of MEK1, had no effect on NO and IL-6 release. Northern blot analysis showed that the p38 MAPK pathway controlled iNOS and IL-6 transcription levels. These data suggest that p38 MAPK plays an important role in the secretion of NO and IL-6 in LPS/IFN- gamma or TNF- alpha /IFN- gamma -treated MC3T3E-1 osteoblasts.


Asunto(s)
Imidazoles/farmacología , Interleucina-6/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Óxido Nítrico Sintasa/antagonistas & inhibidores , Osteoblastos/enzimología , Piridinas/farmacología , Animales , Células Cultivadas , Citocinas/metabolismo , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Expresión Génica/efectos de los fármacos , Interleucina-6/biosíntesis , Interleucina-6/genética , Interleucina-6/metabolismo , Ratones , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Osteoblastos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
19.
Biol Pharm Bull ; 24(5): 453-60, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11379759

RESUMEN

Nitric oxide (NO) induces apoptotic cell death and cAMP has a significantly protective effect on NO-induced cytotoxicity in human osteoblasts, MG-63 cells. Treatment with S-nitroso-N-acetylpenicillamine (SNAP) (0.6 mM) resulted in genomic DNA fragmentation, characteristic of apoptosis. However, concomitant incubation of the cells with either DBcAMP or forskolin markedly inhibited SNAP-induced apoptosis in a dose-dependent manner. Furthermore, pretreatment of MG-63 cells with H-89 or KT5720, which is known to inhibit cAMP-dependent protein kinase (PKA), abolished the protective effect of DBcAMP and forskolin on SNAP-induced apoptosis. In this study, we explored the involvement of caspases in the regulatory mechanism of SNAP-induced apoptosis by cAMP. Our data show that DBcAMP or forskolin blocked SNAP-induced caspase-3-like cysteine protease activation and that H-89, a PKA inhibitor, reversed the cAMP-induced regulatory effect of caspase-3 like protease. Consistent with the results, cAMP inhibited the proteolytic cleavage of caspase-3, -6, -9 and cytochrome c release to cytoplasm. The inhibition of caspase-3 activation did not block SNAP-induced cytochrome c release to cytoplasm, suggesting that caspase-3 activation may occur downstream of cytochrome c release. In summary, these findings show that the exposure of MG-63 cells to cAMP analogs renders them more resistant to NO-induced damage and suggests the presence of regulatory mechanisms of the cell death pathway by cAMP in which caspase-3, -6, and -9 and cytochrome c release serves to mediate NO-induced apoptosis.


Asunto(s)
Apoptosis , Caspasas/fisiología , AMP Cíclico/fisiología , Grupo Citocromo c/metabolismo , Óxido Nítrico/fisiología , Osteoblastos/fisiología , Caspasa 3 , Caspasa 6 , Caspasa 9 , Línea Celular , Colforsina/farmacología , Humanos , Osteoblastos/enzimología , Penicilamina/análogos & derivados , Penicilamina/farmacología
20.
J Leukoc Biol ; 69(3): 490-6, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11261798

RESUMEN

The tumor necrosis factor receptor (TNFR)-associated factor (TRAF) proteins play a central role in the early steps of signal transduction by TNFR superfamily proteins, which induce various cellular responses, including apoptosis. Influences of TRAF proteins on the regulation of cell death and physical interactions between TRAFs and caspases have been reported. In this study, we demonstrate that TRAF3 is proteolyzed during cell death in a caspase-dependent manner. TRAF3 was found to be cleaved by incubation with caspase3 in vitro and by Fas- or CD3-triggering in Jurkat-T cells. The Fas- or CD3-induced cleavage of TRAF3 was blocked by caspase inhibitors and by introduction of alanine substitutions for D347 and D367 residues. Furthermore, the amino-terminal fragment of TRAF3 showed a different intracellular localization from the full-length TRAF3 with preferential distribution to particulate fractions and the nucleus. These findings suggest that TRAF3 may be regulated by caspases during apoptosis of T cells.


Asunto(s)
Caspasas/metabolismo , Células Jurkat/metabolismo , Glicoproteínas de Membrana/farmacología , Proteínas/metabolismo , Clorometilcetonas de Aminoácidos/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Células COS/enzimología , Células COS/metabolismo , Caspasa 3 , Inhibidores de Caspasas , Inhibidores de Cisteína Proteinasa , Proteína Ligando Fas , Células HeLa , Humanos , Células Jurkat/efectos de los fármacos , Células Jurkat/enzimología , Muromonab-CD3/farmacología , Oligopéptidos/farmacología , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Factor 3 Asociado a Receptor de TNF
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA