Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Pers Med ; 14(2)2024 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-38392624

RESUMO

Hypodontia, i.e., missing one or more teeth, is a relatively common human disease; however, oligodontia, i.e., missing six or more teeth, excluding the third molars, is a rare congenital disorder. Many genes have been shown to cause oligodontia in non-syndromic or syndromic conditions. In this study, we identified two novel PAX9 mutations in two non-syndromic oligodontia families. A mutational analysis identified a silent mutation (NM_006194.4: c.771G>A, p.(Gln257=)) in family 1 and a frameshift mutation caused by a single nucleotide duplication (c.637dup, p.(Asp213Glyfs*104)) in family 2. A minigene splicing assay revealed that the silent mutation resulted in aberrant pre-mRNA splicing instead of normal splicing. The altered splicing products are ones with an exon 4 deletion or using a cryptic 5' splicing site in exon 4. Mutational effects were further investigated using protein expression, luciferase activity assay and immunolocalization. We believe this study will not only expand the mutational spectrum of PAX9 mutations in oligodontia but also strengthen the diagnostic power related to the identified silent mutation.

3.
J Pers Med ; 11(6)2021 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-34201399

RESUMO

Hereditary dentin defects can be categorized as a syndromic form predominantly related to osteogenesis imperfecta (OI) or isolated forms without other non-oral phenotypes. Mutations in the gene encoding dentin sialophosphoprotein (DSPP) have been identified to cause dentinogenesis imperfecta (DGI) Types II and III and dentin dysplasia (DD) Type II. While DGI Type I is an OI-related syndromic phenotype caused mostly by monoallelic mutations in the genes encoding collagen type I alpha 1 chain (COL1A1) and collagen type I alpha 2 chain (COL1A2). In this study, we recruited families with non-syndromic dentin defects and performed candidate gene sequencing for DSPP exons and exon/intron boundaries. Three unrelated Korean families were further analyzed by whole-exome sequencing due to the lack of the DSPP mutation, and heterozygous COL1A2 mutations were identified: c.3233G>A, p.(Gly1078Asp) in Family 1 and c.1171G>A, p.(Gly391Ser) in Family 2 and 3. Haplotype analysis revealed different disease alleles in Families 2 and 3, suggesting a mutational hotspot. We suggest expanding the molecular genetic etiology to include COL1A2 for isolated dentin defects in addition to DSPP.

4.
Exp Mol Med ; 52(3): 514-528, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32210358

RESUMO

Osteoclasts (OCs), cells specialized for bone resorption, are generated from monocyte/macrophage precursors by a differentiation process governed by RANKL. Here, we show that DCTN1, a key component of the dynactin complex, plays important roles in OC differentiation. The expression of DCTN1 was upregulated by RANKL. The inhibition of DCTN1 expression by gene knockdown suppressed OC formation, bone resorption, and the induction of NFATc1 and c-Fos, critical transcription factors for osteoclastogenesis. More importantly, the activation of Cdc42 by RANKL was inhibited upon DCTN1 silencing. The forced expression of constitutively active Cdc42 restored the OC differentiation of precursors with DCTN1 deletion. In addition, PAK2 was found to be activated by RANKL and to function downstream of Cdc42. The DCTN1-Cdc42 axis also inhibited apoptosis and caspase-3 activation. Furthermore, the anti-osteoclastogenic effect of DCTN1 knockdown was verified in an animal model of bone erosion. Intriguingly, DCTN1 overexpression was also detrimental to OC differentiation, suggesting that DCTN1 should be regulated at the appropriate level for effective osteoclastogenesis. Collectively, our results reveal that DCTN1 participates in the activation of Cdc42/PAK2 signaling and the inhibition of apoptosis during osteoclastogenesis.


Assuntos
Complexo Dinactina/metabolismo , Osteoclastos/metabolismo , Osteogênese/fisiologia , Transdução de Sinais/fisiologia , Proteína cdc42 de Ligação ao GTP/metabolismo , Quinases Ativadas por p21/metabolismo , Animais , Apoptose/fisiologia , Reabsorção Óssea/metabolismo , Caspase 3/metabolismo , Diferenciação Celular/fisiologia , Modelos Animais de Doenças , Feminino , Macrófagos/metabolismo , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos ICR , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ligante RANK/metabolismo , Regulação para Cima/fisiologia
5.
J Bone Miner Res ; 35(1): 155-166, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31505052

RESUMO

Metastasis to bone is a frequent occurrence in patients with breast and prostate cancers and inevitably threatens the patient's quality of life and survival. Identification of cancer-derived mediators of bone metastasis and osteolysis may lead to novel therapeutic strategies. In this study, we established highly bone-metastatic PC3 prostate and MDA-MB-231 (MDA) breast cancer cell sublines by in vivo selection in mice. In bone-metastatic cancer cells, the expression and secretion of connective tissue growth factor (CTGF) were highly upregulated. CTGF knockdown in bone-metastatic cells decreased invasion activity and MMP expression. RUNX2 overexpression in the CTGF knockdown cells restored the invasion activity and MMP expression. In addition, CTGF increased RUNX2 protein stability by inducing its acetylation via p300 acetyl transferase. The integrin αvß3 receptor mediated these effects of CTGF. Furthermore, CTGF promoted RUNX2 recruitment to the RANKL promoter, resulting in increased RANKL production from the tumor cells and subsequent stimulation of osteoclastogenesis from precursor cells. In addition, animal model with injection of CTGF knocked-down prostate cancer cells into 6-week old BALB/c male mice showed reduced osteolytic lesions. More importantly, the expression levels of CTGF and RANKL showed a strong positive correlation in human primary breast tumor tissues and were higher in bone metastases than in other site metastases. These findings indicate that CTGF plays crucial roles for osteolytic bone metastasis both by enhancing invasiveness of tumor cells and by producing RANKL for osteoclastogenesis. Targeting CTGF may lead to the development of effective preventive and therapeutic strategies for osteolytic metastasis. © 2019 American Society for Bone and Mineral Research.


Assuntos
Neoplasias Ósseas , Neoplasias da Mama , Neoplasias da Próstata , Animais , Neoplasias Ósseas/genética , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Fator de Crescimento do Tecido Conjuntivo/genética , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Osteoclastos , Neoplasias da Próstata/genética , Qualidade de Vida , Ligante RANK
6.
Mol Cells ; 42(11): 810-819, 2019 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-31707778

RESUMO

For physiological or pathological understanding of bone disease caused by abnormal behavior of osteoclasts (OCs), functional studies of molecules that regulate the generation and action of OCs are required. In a microarray approach, we found the suppression of tumorigenicity 5 (ST5) gene is upregulated by receptor activator of nuclear factor-κB ligand (RANKL), the OC differentiation factor. Although the roles of ST5 in cancer and ß-cells have been reported, the function of ST5 in bone cells has not yet been investigated. Knockdown of ST5 by siRNA reduced OC differentiation from primary precursors. Moreover, ST5 downregulation decreased expression of NFATc1, a key transcription factor for osteoclastogenesis. In contrast, overexpression of ST5 resulted in the opposite phenotype of ST5 knockdown. In immunocytochemistry experiments, the ST5 protein is colocalized with Src in RANKL-committed cells. In addition, ST5 enhanced activation of Src and Syk, a Src substrate, in response to RANKL. ST5 reduction caused a decrease in RANKL-evoked calcium oscillation and inhibited translocation of NFATc1 into the nucleus. Taken together, these findings provide the first evidence of ST5 involvement in positive regulation of osteoclastogenesis via Src/Syk/calcium signaling.


Assuntos
Sinalização do Cálcio/genética , Proteínas de Ligação a DNA/genética , Osteoclastos/metabolismo , Osteogênese/genética , Quinase Syk/genética , Proteínas Supressoras de Tumor/genética , Quinases da Família src/genética , Animais , Reabsorção Óssea/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos Endogâmicos ICR , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/citologia , Osteogênese/efeitos dos fármacos , Ligante RANK/farmacologia , Interferência de RNA , Quinase Syk/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Quinases da Família src/metabolismo
7.
Bone Res ; 7: 30, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31667000

RESUMO

Bone destruction induced by breast cancer metastasis causes severe complications, including death, in breast cancer patients. Communication between cancer cells and skeletal cells in metastatic bone microenvironments is a principal element that drives tumor progression and osteolysis. Tumor-derived factors play fundamental roles in this form of communication. To identify soluble factors released from cancer cells in bone metastasis, we established a highly bone-metastatic subline of MDA-MB-231 breast cancer cells. This subline (mtMDA) showed a markedly elevated ability to secrete S100A4 protein, which directly stimulated osteoclast formation via surface receptor RAGE. Recombinant S100A4 stimulated osteoclastogenesis in vitro and bone loss in vivo. Conditioned medium from mtMDA cells in which S100A4 was knocked down had a reduced ability to stimulate osteoclasts. Furthermore, the S100A4 knockdown cells elicited less bone destruction in mice than the control knockdown cells. In addition, administration of an anti-S100A4 monoclonal antibody (mAb) that we developed attenuated the stimulation of osteoclastogenesis and bone loss by mtMDA in mice. Taken together, our results suggest that S100A4 released from breast cancer cells is an important player in the osteolysis caused by breast cancer bone metastasis.

8.
Cell Death Dis ; 10(11): 826, 2019 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-31672960

RESUMO

New bone anabolic agents for the effective treatment of bone metabolic diseases like osteoporosis are of high clinical demand. In the present study, we reveal the function of salt-inducible kinase 1 (SIK1) in regulating osteoblast differentiation. Gene knockdown of SIK1 but not of SIK2 or SIK3 expression in primary preosteoblasts increased osteoblast differentiation and bone matrix mineralization. SIK1 also regulated the proliferation of osteoblastic precursor cells in osteogenesis. This negative control of osteoblasts required the catalytic activity of SIK1. SIK1 phosphorylated CREB regulated transcription coactivator 1 (CRTC1), preventing CRTC1 from enhancing CREB transcriptional activity for the expression of osteogenic genes like Id1. Furthermore, SIK1 knockout (KO) mice had higher bone mass, osteoblast number, and bone formation rate versus littermate wild-type (WT) mice. Preosteoblasts from SIK1 KO mice showed more osteoblastogenic potential than did WT cells, whereas osteoclast generation among KO and WT precursors was indifferent. In addition, bone morphogenic protein 2 (BMP2) suppressed both SIK1 expression as well as SIK1 activity by protein kinase A (PKA)-dependent mechanisms to stimulate osteogenesis. Taken together, our results indicate that SIK1 is a key negative regulator of preosteoblast proliferation and osteoblast differentiation and that the repression of SIK1 is crucial for BMP2 signaling for osteogenesis. Therefore, we propose SIK1 to be a useful therapeutic target for the development of bone anabolic strategies.


Assuntos
Anabolizantes/farmacologia , Osteoporose/tratamento farmacológico , Proteínas Serina-Treonina Quinases/genética , Fatores de Transcrição/genética , Animais , Proteína Morfogenética Óssea 2/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Proteína 1 Inibidora de Diferenciação/genética , Camundongos , Camundongos Knockout , Osteoblastos/efeitos dos fármacos , Osteoporose/genética , Osteoporose/patologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores
9.
Biochem Biophys Res Commun ; 516(1): 202-208, 2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31204051

RESUMO

Mitofusin 2 (Mfn2) is a mitochondrial outer membrane protein that participates in tethering mitochondria to the ER. Mitochondria-ER tethering has been demonstrated to play important roles in many cellular activities by regulating homeostasis of metabolites and calcium. Intracellular calcium signaling is crucial for the differentiation of osteoclasts, the bone-resorbing cells. In this study, we investigated whether Mfn2 plays a role in osteoclastogenesis by receptor activator of nuclear factor kappa B (RANKL) in primary cells. We found that RANKL increased Mfn2 expression during osteoclast formation from mouse bone marrow-derived macrophages (BMMs). When Mfn2 expression was suppressed in BMMs by using a siRNA-mediated gene knock-down system, osteoclast differentiation and activity of mature osteoclasts were reduced. Mfn2 knock-down also decreased the RANKL-mediated induction of NFATc1, the key transcription factor for osteoclast gene expression, without affecting c-Fos level. This effect on NFATc1 was associated with decreased calcium oscillation and calcineurin activity in Mfn2-deficient osteoclasts. Taken together, our results indicate that Mfn2 positively contributes to RANKL-induced osteoclast differentiation by regulating the calcium-calcieurin-NFATc1 axis, raising the importance of a previously under-recognized role of mitochondria in osteoclastogenesis.


Assuntos
Calcineurina/metabolismo , Cálcio/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Fatores de Transcrição NFATC/metabolismo , Osteogênese , Transdução de Sinais , Animais , Sinalização do Cálcio , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos ICR , Mitocôndrias/metabolismo , Osteoclastos/citologia , Osteoclastos/metabolismo
10.
BMB Rep ; 52(11): 659-664, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31072447

RESUMO

Vav1 is a Rho/Rac guanine nucleotide exchange factor primarily expressed in hematopoietic cells. In this study, we investigated the potential role of Vav1 in osteoclast (OC) differentiation by comparing the ability of bone marrow mononuclear cells (BMMCs) obtained from Vav1-deficient (Vav1-/-) and wild-type (WT) mice to differentiate into mature OCs upon stimulation with macrophage colony stimulating factor and receptor activator of nuclear kappa B ligand in vitro. Our results suggested that Vav1 deficiency promoted the differentiation of BMMCs into OCs, as indicated by the increased expression of tartrate-resistant acid phosphatase, cathepsin K, and calcitonin receptor. Therefore, Vav1 may play a negative role in OC differentiation. This hypothesis was supported by the observation of more OCs in the femurs of Vav1-/- mice than in WT mice. Furthermore, the bone status of Vav1-/- mice was analyzed in situ and the femurs of Vav1-/- mice appeared abnormal, with poor bone density and fewer number of trabeculae. In addition, Vav1-deficient OCs showed stronger adhesion to vitronectin, an αvß3 integrin ligand important in bone resorption. Thus, Vav1 may inhibit OC differentiation and protect against bone resorption. [BMB Reports 2019; 52(11): 659-664].


Assuntos
Osteoclastos/metabolismo , Proteínas Proto-Oncogênicas c-vav/metabolismo , Animais , Células da Medula Óssea/metabolismo , Reabsorção Óssea/metabolismo , Osso e Ossos/metabolismo , Diferenciação Celular/fisiologia , Integrina alfaVbeta3/metabolismo , Fator Estimulador de Colônias de Macrófagos/metabolismo , Fator Estimulador de Colônias de Macrófagos/farmacologia , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Osteoclastos/citologia , Proteínas Proto-Oncogênicas c-vav/genética , Proteínas Proto-Oncogênicas c-vav/fisiologia , Ligante RANK/metabolismo , Ligante RANK/fisiologia , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Fosfatase Ácida Resistente a Tartarato
11.
J Immunol ; 202(12): 3359-3369, 2019 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-31076532

RESUMO

Haptoglobin (Hp), a type of acute-phase protein, is known to have a systemic anti-inflammatory function and to modulate inflammation by directly affecting immune cells, such as T cells, dendritic cells, and macrophages. However, the effects of Hp on osteoclast differentiation are not well studied, even though osteoclast precursor cells belong to a macrophage-monocyte lineage. In this study, we found that the bone volume was reduced, and the number of osteoclasts was increased in Hp-deficient mice compared with wild-type mice. Moreover, our in vitro studies showed that Hp inhibits osteoclastogenesis by reducing the protein level of c-Fos at the early phase of osteoclast differentiation. We revealed that Hp-induced suppression of c-Fos was mediated by increased IFN-ß levels. Furthermore, Hp stimulated IFN-ß via a TLR4-dependent mechanism. These results demonstrate that Hp plays a protective role against excessive osteoclastogenesis via the Hp-TLR4-IFN-ß axis.


Assuntos
Haptoglobinas/metabolismo , Interferon beta/metabolismo , Osteoclastos/fisiologia , Reação de Fase Aguda , Animais , Reabsorção Óssea/genética , Diferenciação Celular , Células Cultivadas , Haptoglobinas/genética , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteogênese , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Transdução de Sinais
12.
Materials (Basel) ; 12(5)2019 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-30836587

RESUMO

Zirconia is worth studying as an alternative to dental titanium implants to overcome the disadvantages of titanium. This study investigated the surface characteristics of the zirconia implant material and osteoclastogenesis responses on the surface compared with titanium. Yttrium oxide-stabilized 5% tetragonal zirconia polycrystalline specimens were manufactured, and osteoclast-precursor cells were cultured and differentiated into osteoclasts on the specimens. Surface shape, roughness, and chemical composition were evaluated. After culturing, cell morphologies and differentiation capacity were analyzed using tartrate-resistant acid phosphatase activity (TRACP). mRNA of two critical transcription factors, nuclear factor of activated T-cells 1 (NFATc1) and c-Fos were measured, and protein levels of NFATc1 and c-Fos were investigated. The zirconia specimens had rhomboid-like shapes with smooth surfaces and exhibited no difference in surface roughness compared to the titanium specimens. Morphologies of differentiated osteoclasts on both materials were similar. TRACP activity on the zirconia showed comparable results to that on the titanium. The mRNA value of NFATc1 on the zirconia was higher than that on the titanium at day four. The protein level of c-Fos was expressed thicker on the zirconia when compared to the titanium at day two. The results of this study suggest that zirconia material provides adequate osteoclastogenesis behaviors for dental implant use.

13.
J Bone Metab ; 26(1): 1, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30899717
14.
Clin Oral Investig ; 23(3): 1481-1487, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30120606

RESUMO

OBJECTIVE: Amelogenesis imperfecta (AI) is a rare hereditary disorder affecting the quality and quantity of the tooth enamel. The purpose of this study was to identify the genetic etiology of hypoplastic AI families based on the candidate gene approach. MATERIALS AND METHODS: We recruited three Turkish families with hypoplastic AI and performed a candidate gene screening based on the characteristic clinical feature to find the pathogenic genetic etiology. RESULTS: The candidate gene sequencing of the LAMB3 gene for family 1 revealed a heterozygous nonsense mutation in the last exon [c.3431C > A, p.(Ser1144*)]. FAM20A gene sequencing for families 2 and 3 identified a homozygous deletion [c.34_35delCT, p.(Leu12Alafs*67)] and a homozygous deletion-insertion (c.1109 + 3_1109 + 7delinsTGGTC) mutation, respectively. CONCLUSION: The candidate gene approach can be successfully used to identify the genetic etiology of the AI in some cases with characteristic clinical features. CLINICAL RELEVANCE: Identification of the genetic etiology of the AI will help both the family members and dentist understand the nature of the disorder. Characteristic clinical feature can suggest possible genetic causes.


Assuntos
Amelogênese Imperfeita/genética , Moléculas de Adesão Celular/genética , Proteínas do Esmalte Dentário/genética , Códon sem Sentido , Análise Mutacional de DNA , Homozigoto , Humanos , Mutação INDEL , Linhagem , Deleção de Sequência , Turquia , Calinina
15.
Data Brief ; 18: 518-522, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29900211

RESUMO

C-X-C motif chemokine receptor 3 (CXCR3) is a G protein-coupled receptor for three ligands which are C-X-C motif chemokine 9 (CXCL9), CXCL10, and CXCL11 [1]. Previously we have reported that CXCL10 promotes pro-inflammatory cytokine expression, and forms positive feedback loop [2], [3]. In the present study, we described mRNA expression of CXCL9 and CXCL11 under CXCL10 stimuli in the presence or absence of CXCR3 antagonist, JN-2 in bone marrow-derived macrophages (BMMs) and CD4+ T cells. In addition, we examined pro-inflammatory cytokine expression under CXCL9 or CXCL11 stimuli in BMMs and CD4+ T cells.

16.
Cell Signal ; 49: 68-78, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29879471

RESUMO

Platelet-derived growth factor receptor (PDGFR) signaling has been shown to inhibit osteogenesis. However, therapeutic efficacy of inhibiting PDGF signaling to bone regeneration in vivo and the specific mechanisms by which PDGFR signaling inhibits osteogenic differentiation remain unclear. In the present study, we examined the osteogenic effect of inhibiting PDGFR using trapidil, a PDGFR antagonist, in vivo and in vitro, and evaluated its mechanisms. A rat calvarial defect model was analyzed by micro-computed tomography and histology to determine the pro-osteogenic effect of trapidil in vivo. In addition, primary mouse calvarial osteoblast precursors were cultured in osteogenic differentiation medium with trapidil to study the mechanisms. Trapidil greatly promoted bone regeneration in a rat calvarial defect model and osteogenic differentiation of calvarial osteoblast precursors. For the mechanisms, trapidil induced phosphorylation of Smad1/5/9 and mitogen-activated protein kinase (MAPK) leading to enhance expression of Runx2, crucial transcription factor for osteogenesis. The pro-osteogenic effects of trapidil were inhibited by LDN193189, specific inhibitor of bone morphogenetic protein (BMP) receptor, ALK2 and ALK3, and by depletion of ALK3, and treatment with noggin, an antagonist of BMPs. Moreover, trapidil showed a synergistic effect with BMP2 on osteogenic differentiation. In conclusion, trapidil induced BMPR activity through upregulation of BMP signaling, leading to promoted osteogenesis in vitro and in vivo. Attenuated BMPR activity may be involved in the inhibition of osteogenesis by PDGFR signaling.


Assuntos
Osteogênese/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Trapidil/farmacologia , Regulação para Cima/efeitos dos fármacos , Fosfatase Alcalina/antagonistas & inibidores , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Transporte/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Masculino , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Crânio/citologia , Proteínas Smad/genética , Proteínas Smad/metabolismo
17.
J Bone Metab ; 25(1): 23-33, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29564303

RESUMO

BACKGROUND: Breast cancer is one of the most common cancers affecting women and has a high incidence of bone metastasis, causing osteolytic lesions. The elevated expression of receptor activator of nuclear factor-κB ligand (RANKL) in cancer activates osteoclasts, leading to bone destruction. We previously reported that α-tocopheryl succinate (αTP-suc) inhibited interleukin-1-induced RANKL expression in osteoblasts. Here, we examined the effect of αTP-suc on osteolytic bone metastasis in breast cancer. METHODS: To examine the effect of αTP-suc on the metastatic capacity of breast cancer, MDA-MB-231-FL cells were injected into the left cardiac ventricle of BALB/c nude mice along with intraperitoneal injection of αTP-suc. The mice were then analyzed by bioluminescence imaging. To investigate the effect of αTP-suc on osteolysis, 4T1 cells were directly injected into the femur of BALB/c mice along with intraperitoneal injection of αTP-suc. Microcomputed tomography analysis and histomorphometric analysis of the femora were performed. RESULTS: αTP-suc inhibited cell migration and cell growth of 4T1 cells. In line with these results, bone metastasis of MDA-MB-231-FL cells was reduced in mice injected with αTP-suc. In addition, αTP-suc decreased osteoclastogenesis by inhibiting 4T1-induced RANKL expression in osteoblasts. Consistent with these results, 4T1-induced bone destruction was ameliorated by αTP-suc, with in vivo analysis showing reduced tumor burden and osteoclast numbers. CONCLUSIONS: Our findings suggest that αTP-suc may be efficiently utilized to prevent and treat osteolytic bone metastasis of breast cancer with dual effects.

18.
Eur J Pharmacol ; 823: 1-10, 2018 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-29378189

RESUMO

Rheumatoid arthritis (RA) is a chronic autoimmune disease that is characterized by uncontrolled joint inflammation and destruction of bone and cartilage. Previous studies have shown that C-X-C motif chemokine 10 (CXCL10) has important roles in RA development and that blocking CXCL10 expression effectively inhibits arthritis progression in animal models. However, clinical study using anti-CXCL10 monoclonal antibody (MDX-1100) to block CXCL10 expression in patients with RA did not show significant effectiveness. Therefore, we turned our attention to C-X-C motif chemokine receptor 3 (CXCR3), which is a receptor for CXCL9, CXCL10, and CXCL11, to treat RA. In the present study, administration of JN-2, our newly developed CXCR3 antagonist, ameliorated the progression of arthritis in a collagen-induced arthritis animal model. JN-2 also inhibited CXCR3-induced cell migration and pro-inflammatory cytokine expression of bone marrow-derived macrophages and CD4+ T cells in vitro. In addition, we found that CXCL10 formed an auto-amplification loop through activation of NFκB. Furthermore, Phosphorylation of p65 at serine 536 played an important role in the auto-amplification of CXCL10. Overall, the present results demonstrated that JN-2 decreased inflammation by inhibiting CXCR3-enhanced cell migration and pro-inflammatory cytokine expression, which then ameliorated arthritis progression.


Assuntos
Artrite Reumatoide/tratamento farmacológico , Quimiocinas CXC/antagonistas & inibidores , Progressão da Doença , Oxazóis/farmacologia , Animais , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Movimento Celular/efeitos dos fármacos , Quimiocina CXCL10/metabolismo , Citocinas/biossíntese , Modelos Animais de Doenças , Masculino , Camundongos , Oxazóis/uso terapêutico , Fator de Transcrição RelA/metabolismo
19.
Sci Rep ; 7(1): 10626, 2017 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-28878266

RESUMO

Transglutaminase 2 (TG2) performs multiple reactions, including transamidation, and also plays a role in signal transduction as a GTP-binding protein. In this study, we reveal that TG2 controls osteoclast differentiation and bone homeostasis in mice. Osteoclasts specifically expressed the TG2 isoform among eight TG family members. Suppression in TG2 expression with siRNA led to increased osteoclast formation from primary mouse precursor cells in response to receptor activator of nuclear factor kappaB ligand (RANKL). This osteoclastogenic effect of TG2 knockdown was associated with enhanced induction of c-Fos and NFATc1 by RANKL. Moreover, TG2 knockdown up-regulated B lymphocyte-induced maturation protein 1 (Blimp1), which represses anti-osteoclastogenic genes, in a manner dependent on the NF-κB signaling pathway. To the contrary, TG2 overexpression inhibited osteoclast formation and the expression of osteoclastogenic genes. Consistent with these in vitro results, TG2 knockout mice exhibited lower trabecular bone mass and increased number of osteoclasts compared with wild-type mice. Taken together, our results provide strong evidence that TG2 plays an important role in bone metabolism by suppressing excessive osteoclastogenesis via the regulation of the NF-κB-Blimp1 signaling pathway.


Assuntos
Diferenciação Celular , Proteínas de Ligação ao GTP/metabolismo , Osteoclastos/citologia , Osteoclastos/metabolismo , Fator 1 de Ligação ao Domínio I Regulador Positivo/metabolismo , Transdução de Sinais , Transglutaminases/metabolismo , Animais , Densidade Óssea/genética , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Diferenciação Celular/genética , Linhagem Celular , Proteínas de Ligação ao GTP/genética , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , Modelos Biológicos , NF-kappa B/metabolismo , Fator 1 de Ligação ao Domínio I Regulador Positivo/genética , Proteína 2 Glutamina gama-Glutamiltransferase , Ligante RANK/metabolismo , Ligante RANK/farmacologia , Transglutaminases/genética , Microtomografia por Raio-X
20.
Exp Mol Med ; 49(7): e358, 2017 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-28729639

RESUMO

Osteoclasts are responsible for the bone erosion associated with rheumatoid arthritis (RA). The upregulation of the chemokines CCL19 and CCL21 and their receptor CCR7 has been linked to RA pathogenesis. The purpose of this study was to evaluate the effects of CCL19 and CCL21 on osteoclasts and to reveal their underlying mechanisms. The expression of CCL19, CCL21 and CCR7 was higher in RA patients than in osteoarthritis patients. In differentiating osteoclasts, tumor necrosis factor-α, interleukin-1ß and lipopolysaccharide stimulated CCR7 expression. CCL19 and CCL21 promoted osteoclast migration and resorption activity. These effects were dependent on the presence of CCR7 and abolished by the inhibition of the Rho signaling pathway. CCL19 and CCL21 promoted bone resorption by osteoclasts in an in vivo mice calvarial model. These findings demonstrate for the first time that CCL19, CCL21 and CCR7 play important roles in bone destruction by increasing osteoclast migration and resorption activity. This study also suggests that the interaction of CCL19 and CCL21 with CCR7 is an effective strategic focus in developing therapeutics for alleviating inflammatory bone destruction.


Assuntos
Reabsorção Óssea/metabolismo , Movimento Celular , Quimiocina CCL19/metabolismo , Quimiocina CCL21/metabolismo , Osteoclastos/metabolismo , Receptores CCR7/metabolismo , Animais , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Diferenciação Celular , Quimiocina CCL19/sangue , Quimiocina CCL21/sangue , Citocinas/metabolismo , Feminino , Humanos , Ligantes , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos ICR , Osteoartrite/metabolismo , Osteoartrite/patologia , Proteína rhoA de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...