Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 279
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Cell Biochem Funct ; 42(4): e4068, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38817105

RESUMO

Evidence is accumulating that osteal macrophages, in addition to bone-resorbing osteoclasts and bone-forming osteoblasts, participate vitally in bone remodeling process. Oncostatin M (OSM), an inflammatory cytokine belonging to interleukin-6 superfamily, is recognized as an essential factor secreted by osteal macrophages to orchestrate bone remodeling. Osteoprotegerin (OPG) produced by osteoblasts regulates osteoclastogenesis. We have reported that bone morphogenetic protein-4 (BMP-4) stimulates OPG synthesis in MC3T3-E1 osteoblast-like cells, and that SMAD1/5/8(9), p38 mitogen-activated protein kinase (MAPK), and p70 S6 kinase are involved in the OPG synthesis. The present study aims to investigate the effect of OSM on the synthesis of OPG stimulated by BMP-4 in osteoblasts. OSM suppressed the release and the mRNA expression of OPG upregulated by BMP-4 in MC3T3-E1 cells. Neither the BMP-4-induced phosphorylation of SMAD1/5/9 nor that of p38 MAPK was affected by OSM. On the other hand, the phosphorylation of p70 S6 kinase stimulated by BMP-4 was considerably suppressed by OSM. These results strongly suggest that OSM suppresses the BMP-4-stimulated OPG synthesis via inhibition of the p70 S6 kinase-mediated pathway in osteoblast-like cells.


Assuntos
Proteína Morfogenética Óssea 4 , Oncostatina M , Osteoblastos , Osteoprotegerina , Proteínas Quinases S6 Ribossômicas 70-kDa , Animais , Camundongos , Oncostatina M/farmacologia , Oncostatina M/metabolismo , Osteoblastos/metabolismo , Osteoblastos/efeitos dos fármacos , Osteoblastos/citologia , Osteoprotegerina/metabolismo , Osteoprotegerina/biossíntese , Proteína Morfogenética Óssea 4/metabolismo , Proteína Morfogenética Óssea 4/farmacologia , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Linhagem Celular
2.
Cell Physiol Biochem ; 56(1): 28-38, 2022 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-35060690

RESUMO

BACKGROUND/AIMS: Osteoprotegerin (OPG) is a profibrotic mediator produced by myofibro-blasts under influence of transforming growth factor ß (TGFß). Its expression in experimental models of liver fibrosis correlates well with disease severity and treatment responses. The regulation of OPG in liver tissue is largely unknown and we therefore set out to elucidate which growth factors/interleukins associated with fibrosis induce OPG and through which pathways. METHODS: Precision-cut liver slices of wild type and STAT6-deficient mice and 3T3 fibroblasts were used to investigate the effects of TGFß, interleukin (IL) 13 (IL13), IL1ß, and platelet-derived growth factor BB (PDGF-BB) on expression of OPG. OPG protein was measure by ELISA, whereas OPG mRNA and expression of other relevant genes was measured by qPCR. RESULTS: In addition to TGFß, only IL13 and not PDGF-BB or IL1ß could induce OPG expression in 3T3 fibroblasts and liver slices. This IL13-dependent induction was not shown in liver slices of STAT6-deficient mice and when wild type slices were cotreated with TGFß receptor 1 kinase inhibitor galunisertib, STAT6 inhibitor AS1517499, or AP1 inhibitor T5224. This suggests that the OPG-inducing effect of IL13 is mediated through IL13 receptor α1-activation and subsequent STAT6-dependent upregulation of IL13 receptor α2, which in turn activates AP1 and induces production of TGFß and subsequent production of OPG. CONCLUSION: We have shown that IL13 induces OPG release by liver tissue through a TGFß-dependent pathway involving both the α1 and the α2 receptor of IL13 and transcription factors STAT6 and AP1. OPG may therefore be a novel target for the treatment liver fibrosis as it is mechanistically linked to two important regulators of fibrosis in liver, namely IL13 and TGFß1.


Assuntos
Regulação da Expressão Gênica , Interleucina-13/metabolismo , Cirrose Hepática/metabolismo , Fígado/metabolismo , Osteoprotegerina/biossíntese , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Feminino , Masculino , Camundongos
3.
Horm Metab Res ; 54(1): 42-49, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34986499

RESUMO

Incretins including glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1), which are secreted from the small intestine after oral food ingestion, are currently well-known to stimulate insulin secretion from pancreatic ß-cells and used for the treatment of type 2 diabetes mellitus. We have previously reported that prostaglandin F2α (PGF2α) stimulates the synthesis of interleukin-6 (IL-6) and osteoprotegerin in osteoblast-like MC3T3-E1 cells, and that IL-6 and osteoprotegerin release are mediated through the p44/p42 mitogen-activated protein (MAP) kinase, p38 MAP kinase or stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) pathways. In the present study, we investigated the effects of incretins including GLP-1 and GIP, on the PGF2α-induced synthesis of IL-6 and osteoprotegerin and examined the detailed mechanism in osteoblast-like MC3T3-E1 cells. We found that GIP and GLP-1 significantly stimulated the PGF2α-induced synthesis of IL-6 in osteoblast-like MC3T3-E1 cells. In addition, GIP and GLP-1 significantly enhanced the PGF2α-induced mRNA expression levels of IL-6. On the other hand, GIP and GLP-1 markedly stimulated the PGF2α-induced synthesis of osteoprotegerin. However, the phosphorylation of p44/p42 MAP kinase, p38 MAP kinase, or JNK induced by PGF2α was not affected by GIP or GLP-1. Therefore, these results strongly suggest that incretins enhance the PGF2α-induced synthesis of IL-6 and osteoprotegerin in osteoblast-like MC3T3-E1 cells. However, these syntheses are not mediated through p44/p42 MAP kinase, p38 MAP kinase, or JNK pathways.


Assuntos
Dinoprosta/farmacologia , Polipeptídeo Inibidor Gástrico/farmacologia , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Incretinas/metabolismo , Interleucina-6/biossíntese , Osteoblastos/metabolismo , Osteoprotegerina/biossíntese , Animais , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Interleucina-6/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Osteoblastos/efeitos dos fármacos , Osteoblastos/enzimologia , Fosforilação/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
4.
J Cell Mol Med ; 25(5): 2390-2403, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33511706

RESUMO

Osteoclasts (OCs) differentiate from the monocyte/macrophage lineage, critically regulate bone resorption and remodelling in both homeostasis and pathology. Various immune and non-immune cells help initiating activation of myeloid cells for differentiation, whereas hyper-activation leads to pathogenesis, and mechanisms are yet to be completely understood. Herein, we show the efficacy of dental pulp-derived stem cells (DPSCs) in limiting RAW 264.7 cell differentiation and underlying molecular mechanism, which has the potential for future therapeutic application in bone-related disorders. We found that DPSCs inhibit induced OC differentiation of RAW 264.7 cells when co-cultured in a contact-free system. DPSCs reduced expression of key OC markers, such as NFATc1, cathepsin K, TRAP, RANK and MMP-9 assessed by quantitative RT-PCR, Western blotting and immunofluorescence detection methods. Furthermore, quantitative RT-PCR analysis revealed that DPSCs mediated M2 polarization of RAW 264.7 cells. To define molecular mechanisms, we found that osteoprotegerin (OPG), an OC inhibitory factor, was up-regulated in RAW 264.7 cells in the presence of DPSCs. Moreover, DPSCs also constitutively secrete OPG that contributed in limiting OC differentiation. Finally, the addition of recombinant OPG inhibited OC differentiation in a dose-dependent manner by reducing the expression of OC differentiation markers, NFATc1, cathepsin K, TRAP, RANK and MMP9 in RAW 264.7 cells. RNAKL and M-CSF phosphorylate AKT and activate PI3K-AKT signalling pathway during osteoclast differentiation. We further confirmed that OPG-mediated inhibition of the downstream activation of PI3K-AKT signalling pathway was similar to the DPSC co-culture-mediated inhibition of OC differentiation. This study provides novel evidence of DPSC-mediated inhibition of osteoclastogenesis mechanisms.


Assuntos
Diferenciação Celular , Polpa Dentária/citologia , Osteoclastos/metabolismo , Osteoprotegerina/biossíntese , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Animais , Biomarcadores , Células Cultivadas , Técnicas de Cocultura , Regulação da Expressão Gênica , Humanos , Mediadores da Inflamação , Camundongos , Células Mieloides/citologia , Células Mieloides/metabolismo , Osteoclastos/citologia , Células RAW 264.7 , Células-Tronco/citologia , Estresse Fisiológico
5.
Int J Mol Sci ; 22(9)2021 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-33946862

RESUMO

Lumican, a ubiquitously expressed small leucine-rich proteoglycan, has been utilized in diverse biological functions. Recent experiments demonstrated that lumican stimulates preosteoblast viability and differentiation, leading to bone formation. To further understand the role of lumican in bone metabolism, we investigated its effects on osteoclast biology. Lumican inhibited both osteoclast differentiation and in vitro bone resorption in a dose-dependent manner. Consistent with this, lumican markedly decreased the expression of osteoclastogenesis markers. Moreover, the migration and fusion of preosteoclasts and the resorptive activity per osteoclast were significantly reduced in the presence of lumican, indicating that this protein affects most stages of osteoclastogenesis. Among RANKL-dependent pathways, lumican inhibited Akt but not MAP kinases such as JNK, p38, and ERK. Importantly, co-treatment with an Akt activator almost completely reversed the effect of lumican on osteoclast differentiation. Taken together, our findings revealed that lumican inhibits osteoclastogenesis by suppressing Akt activity. Thus, lumican plays an osteoprotective role by simultaneously increasing bone formation and decreasing bone resorption, suggesting that it represents a dual-action therapeutic target for osteoporosis.


Assuntos
Reabsorção Óssea/fisiopatologia , Lumicana/farmacologia , Osteoclastos/metabolismo , Osteogênese/fisiologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Fusão Celular , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Lumicana/fisiologia , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos ICR , Osteogênese/efeitos dos fármacos , Osteoprotegerina/biossíntese , Ligante RANK/biossíntese , Ligante RANK/farmacologia , Proteínas Recombinantes/farmacologia
6.
J Cell Mol Med ; 24(18): 10542-10550, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32783377

RESUMO

Calcium deposition in vascular smooth muscle cells (VSMCs) is a form of ectopic ossification in blood vessels. It can result in rigidity of the vasculature and an increase in cardiac events. Here, we report that the microRNA miR-134-5p potentiates inorganic phosphate (Pi)-induced calcium deposition in VSMCs by inhibiting histone deacetylase 5 (HDAC5). Using miRNA microarray analysis of Pi-treated rat VSMCs, we first selected miR-134-5p for further evaluation. Quantitative RT-PCR confirmed that miR-134-5p was increased in Pi-treated A10 cells, a rat VSMC line. Transfection of miR-134-5p mimic potentiated the Pi-induced increase in calcium contents. miR-134-5p increased the amounts of bone runt-related transcription factor 2 (RUNX2) protein and bone morphogenic protein 2 (BMP2) mRNA in the presence of Pi but decreased the expression of osteoprotegerin (OPG). Bioinformatic analysis showed that the HDAC5 3'untranslated region (3'UTR) was one of the targets of miR-134-5p. The luciferase construct containing the 3'UTR of HDAC5 was down-regulated by miR-134-5p mimic in a dose-dependent manner in VSMCs. Overexpression of HDAC5 mitigated the calcium deposition induced by miR-134-5p. Our results suggest that a Pi-induced increase of miR-134-5p may cause vascular calcification through repression of HDAC5.


Assuntos
Cálcio/metabolismo , Histona Desacetilases/efeitos dos fármacos , MicroRNAs/fisiologia , Miócitos de Músculo Liso/metabolismo , Calcificação Vascular/etiologia , Regiões 3' não Traduzidas , Animais , Aorta Torácica/citologia , Linhagem Celular , Simulação por Computador , Subunidade alfa 1 de Fator de Ligação ao Core/biossíntese , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/imunologia , Regulação para Baixo , Regulação da Expressão Gênica , Genes Reporter , Histona Desacetilases/biossíntese , Histona Desacetilases/genética , MicroRNAs/genética , Análise em Microsséries , Músculo Liso Vascular/citologia , Osteoprotegerina/biossíntese , Osteoprotegerina/genética , Fosfatos/toxicidade , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transfecção , Calcificação Vascular/genética , Calcificação Vascular/metabolismo , Calcificação Vascular/prevenção & controle
7.
Arterioscler Thromb Vasc Biol ; 39(3): 432-445, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30626205

RESUMO

Objective- Vascular smooth muscle cell (VSMC) transformation to an osteochondrogenic phenotype is an initial step toward arterial calcification, which is highly correlated with cardiovascular disease-related morbidity and mortality. TLR2 (Toll-like receptor 2) plays a pathogenic role in the development of vascular diseases, but its regulation in calcification of arteries and VSMCs remains unclear. We postulate that TLR2-mediated inflammation participates in mediating atherosclerotic arterial calcification and VSMC calcification. Approach and Results- We found that ApoE-/- Tlr2-/- genotype in mice suppressed high-fat diet-induced atherosclerotic plaques formation during initiation but progressively lost its preventative capacity, compared with ApoE-/- mice. However, TLR2 deficiency prohibited high-fat diet-induced advanced atherosclerotic calcification, chondrogenic metaplasia, and OPG (osteoprotegerin) downregulation in the calcified lesions. Incubation of VSMCs in a calcifying medium revealed that TLR2 agonists significantly increased VSMC calcification and chondrogenic differentiation. Furthermore, TLR2 deficiency suppressed TLR2 agonist-mediated VSMC chondrogenic differentiation and consequent calcification, which were triggered via the concerted actions of IL (interleukin)-6-mediated RANKL (receptor activator of nuclear factor κB ligand) induction and OPG suppression. Inhibition experiments with pharmacological inhibitors demonstrated that IL-6-mediated RANKL induction is signaled by p38 and ERK1/2 (extracellular signal-regulated kinase 1/2) pathways, whereas the OPG is suppressed via NF-κB (nuclear factor κB) dependent signaling mediated by ERK1/2. Conclusions- We concluded that on ligand binding, TLR2 activates p38 and ERK1/2 signaling to selectively modulate the upregulation of IL-6-mediated RANKL and downregulation of OPG. These signaling pathways act in concert to induce chondrogenic transdifferentiation of VSMCs, which in turn leads to vascular calcification during the pathogenesis of atherosclerosis.


Assuntos
Aterosclerose/patologia , Calcinose/metabolismo , Calcinose/patologia , Condrogênese/fisiologia , Interleucina-6/fisiologia , Sistema de Sinalização das MAP Quinases , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Osteoprotegerina/biossíntese , Ligante RANK/biossíntese , Receptor 2 Toll-Like/fisiologia , Animais , Doenças da Aorta/etiologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Doenças da Aorta/prevenção & controle , Apolipoproteínas E/deficiência , Aterosclerose/etiologia , Aterosclerose/genética , Aterosclerose/prevenção & controle , Calcinose/genética , Células Cultivadas , Colesterol na Dieta/toxicidade , Dieta Hiperlipídica/efeitos adversos , Gorduras na Dieta/toxicidade , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Osteoprotegerina/genética , Ligante RANK/genética , Distribuição Aleatória
8.
BMC Musculoskelet Disord ; 21(1): 375, 2020 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-32532246

RESUMO

BACKGROUND: Age-dependent male osteoporosis remains a poorly studied medical problem despite its significance. It is estimated that at least 1 of 5 men will suffer from osteoporotic consequences. Given that multiple mechanisms are involved in the process of senescence, much attention has been given to compounds with polymodal actions. To challenge such a health problem, we tested here the therapeutic potential of resveratrol in male osteoporosis. We also studied the possible molecular mechanisms that may underlie resveratrol effects. METHODS: Thirty male Wistar albino rats were used in the present study. Rats were divided (10/group) into: control (3-4 months old weighing 150-200 g receiving vehicle), aged (18-20 months old, weighing 350-400 g and receiving vehicle), and resveratrol treated aged (18-20 months old, weighing 350-400 g and receiving resveratrol 20 mg/kg/day for 6 weeks) groups. Assessment of serum calcium, phosphate, bone specific alkaline phosphatase, inflammatory cytokines, oxidative stress markers, and rat femur gene expression of FoxO1, SIRT1, RANKL and OPG proteins was carried out. Histopathological assessment of different levels of rat femur was also performed. RESULTS: Age-dependent osteoporosis resulted in significant increase in serum levels of phosphate, bone specific alkaline phosphatase, hsCRP, IL-1ß, IL-6, TNF-α, MDA, NO, and RANKL gene expression. However, there was significant decrease in serum level of GSH, and gene expression of FoxO1, SIRT1 and OPG. Osteoporotic changes were seen in femur epiphysis, metaphysis and diaphysis. Resveratrol restored significantly age-dependent osteoporotic changes. CONCLUSION: We concluded that resveratrol can play an important role in the prevention of male osteoporosis. Resveratrol can counter the molecular changes in male osteoporosis via anti-inflammatory, anti-oxidant and gene modifying effects.


Assuntos
Envelhecimento/efeitos dos fármacos , Antioxidantes/uso terapêutico , Osteoporose/tratamento farmacológico , Resveratrol/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Envelhecimento/metabolismo , Animais , Antioxidantes/farmacologia , Densidade Óssea/efeitos dos fármacos , Citocinas/metabolismo , Fêmur/patologia , Masculino , Proteínas do Tecido Nervoso/metabolismo , Osteoporose/metabolismo , Osteoprotegerina/biossíntese , Osteoprotegerina/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ligante RANK/biossíntese , Ligante RANK/efeitos dos fármacos , Ratos , Ratos Wistar , Resveratrol/farmacologia , Sirtuína 1/metabolismo
9.
Int J Mol Sci ; 21(15)2020 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-32722636

RESUMO

Chondroitin sulfate (CS) has antioxidative, anti-inflammatory, anti-osteoarthritic and hypoglycemic effects. However, whether it has antidiabetic osteoporosis effects has not been reported. Therefore, in this study, we established a STZ-induced diabetic rat model; CS (500 mg kg-1 d-1) was orally administrated for eight weeks to study its preventive effects on diabetic osteoporosis. The results showed that eight weeks of CS treatment improved the symptoms of diabetes; the CS-treated group has increased body weight, decreased water or food intake, decreased blood glucose, increased bone-mineral density, repaired bone morphology and decreased femoral osteoclasts and tibia adipocytes numbers. After CS treatment, bone histomorphometric parameters returned to normal, the levels of serum inflammatory cytokines (IL-1ß, IL-6 and TNF-α) decreased significantly, serum SOD, GPX and CAT activities increased and MDA level increased. In the CS-treated group, the levels of serum ALP, CTX-1, TRACP 5b, osteocalcin and RANKL decreased and the serum RUNX 2 and OPG levels increased. Bone immunohistochemistry results showed that CS can effectively increase the expression of OPG and RUNX2 and reduce the expression of RANKL in diabetic rats. All of these indicate that CS could prevent STZ induced diabetic osteoporosis-mainly through decreasing blood glucose, antioxidative stress, anti-inflammation and regulation of OPG/RANKL expression. CS can therefore effectively prevent bone loss caused by diabetes.


Assuntos
Glicemia/metabolismo , Sulfatos de Condroitina/farmacologia , Complicações do Diabetes/prevenção & controle , Diabetes Mellitus Experimental/tratamento farmacológico , Regulação da Expressão Gênica/efeitos dos fármacos , Osteoporose/prevenção & controle , Osteoprotegerina/biossíntese , Estresse Oxidativo/efeitos dos fármacos , Ligante RANK/biossíntese , Animais , Complicações do Diabetes/metabolismo , Diabetes Mellitus Experimental/metabolismo , Feminino , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Osteoporose/etiologia , Osteoporose/metabolismo , Ratos , Ratos Sprague-Dawley
10.
Toxicol Appl Pharmacol ; 343: 62-70, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29477364

RESUMO

Depleted uranium (DU) is widely used in military and civil activities, and bone is the main target organ of chronic DU toxicity. The aim of this study was to evaluate the effects of ghrelin on rats implanted with DU and explore the underlying mechanisms. The results showed that ghrelin could increase the expression of ghrelin receptor in bone tissue, thus alleviate the apoptosis of bone tissue after 3 months of 0.3 g DU embedded in the tibia. Micro-computed tomography examination showed that after DU implantation, the density of cortical bone showed no significant difference, but the trabecular bone decreased in amount, density and connectivity. Ghrelin treatment can significantly reduce the changes caused by DU. Moreover, ghrelin can inhibit the increase of serum tartrate resistant acid phosphatase and the decrease of alkaline phosphatase and osteocalcin. Furthermore, ghrelin can also significantly reduce the receptor activator of nuclear factor κB ligand (RANKL) and phosphorylated p38-MAPK expression, and increase the level of osteoprotegerin (OPG) in tissues after exposure to DU. Based on cell experimental research, p38-MAPK specific agonist can reverse the function of ghrelin, significantly inhibit the level of OPG and increase the level of RANKL. On the contrary, the use of p38-MAPK specific inhibitor or p38-MAPK siRNA can enhance the function of ghrelin. These results suggest that ghrelin may inhibit p38 MAPK activation induced by DU, and increase the OPG/RANKL ratio caused by DU exposure, hence alleviating the bone damage caused by long-term DU exposure.


Assuntos
Densidade Óssea/fisiologia , Citoproteção/fisiologia , Grelina/farmacologia , Osteoprotegerina/biossíntese , Ligante RANK/biossíntese , Urânio/toxicidade , Animais , Densidade Óssea/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Citoproteção/efeitos dos fármacos , Masculino , Camundongos , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoblastos/patologia , Ratos , Ratos Sprague-Dawley , Tíbia/efeitos dos fármacos , Tíbia/metabolismo , Tíbia/patologia
11.
Med Sci Monit ; 24: 5292-5300, 2018 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-30059491

RESUMO

BACKGROUND Osteoprotegerin (OPG) inhibits bone resorption and binds with strong affinity to receptor activator of NF κB ligand (RANKL), thereby preventing RANKL from binding to its receptor RANK. Osteoclasts have documented effects on bone erosion of rheumatoid arthritis (RA). The aim of this study was to examine the role of miR-145-5p in the regulation of RA osteoclast differentiation and bone erosion. MATERIAL AND METHODS Expression of microRNA-145-5p in human peripheral blood mononuclear cells (PBMC) and synovial tissue was assayed by real-time polymerase chain reaction (RT-PCR). OPG, RANK, and RANKL expression in RAW-264.7 cells was examined by RT-PCR and Western blot analysis. Osteoclast formation was detected by tartrate-resistant acid phosphatase (TRAP) staining. The effect of miR-145-5p on predicted target mRNAs was examined by luciferase reporter assays. Collagen-induced arthritis (CIA) was induced by injecting DBA/1 mice with bovine type II collagen (CII), and miR-145-5p agomir was administered by intravenous injection. Morphological changes in the CIA joint were assessed by micro-computed tomography (CT) and histopathology. RESULTS miR-145-5p levels significantly increased in RA PBMC and synovial tissue compared with normal PBMC and osteoarthritis (OA) tissue. After transfection of RAW-264.7 cells with miR-145-5p, RANK and RANKL expression increased significantly, while OPG expression decreased significantly. TRAP staining results showed osteoclast numbers increased. Micro-CT analysis of the arthritic joints showed that the miR-145-5p agomir caused bone erosion in mice, and histopathological analysis revealed that miR-145-5p agomir aggravates cartilage erosion. CONCLUSIONS Our findings indicate that administration of miR-145-5p aggravates joint erosion in CIA mice. This suggests that miR-145-5p is a potential target for the treatment of RA.


Assuntos
Artrite Experimental/genética , Reabsorção Óssea/genética , MicroRNAs/biossíntese , Osteoclastos/patologia , Osteoprotegerina/biossíntese , Adulto , Animais , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Artrite Reumatoide/genética , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Reabsorção Óssea/patologia , Bovinos , Colágeno Tipo II/administração & dosagem , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos DBA , MicroRNAs/administração & dosagem , MicroRNAs/sangue , MicroRNAs/genética , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Osteoclastos/metabolismo , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Ligante RANK/biossíntese , Ligante RANK/metabolismo , Células RAW 264.7 , Receptor Ativador de Fator Nuclear kappa-B/biossíntese , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia , Transfecção , Adulto Jovem
12.
Int J Mol Sci ; 19(12)2018 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-30562925

RESUMO

Butyric acid as a histone deacetylase (HDAC) inhibitor is produced by a number of periodontal and root canal microorganisms (such as Porphyromonas, Fusobacterium, etc.). Butyric acid may affect the biological activities of periodontal/periapical cells such as osteoblasts, periodontal ligament cells, etc., and thus affect periodontal/periapical tissue destruction and healing. The purposes of this study were to study the toxic effects of butyrate on the matrix and mineralization marker expression in MG-63 osteoblasts. Cell viability was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. Cellular apoptosis and necrosis were analyzed by propidium iodide/annexin V flow cytometry. The protein and mRNA expression of osteoprotegerin (OPG) and receptor activator of nuclear factor kappa-B ligand (RANKL) were analyzed by Western blotting and reverse transcriptase-polymerase chain reaction (RT-PCR). OPG, soluble RANKL (sRANKL), 8-isoprostane, pro-collagen I, matrix metalloproteinase-2 (MMP-2), osteonectin (SPARC), osteocalcin and osteopontin (OPN) secretion into culture medium were measured by enzyme-linked immunosorbant assay. Alkaline phosphatase (ALP) activity was checked by ALP staining. Histone H3 acetylation levels were evaluated by immunofluorescent staining (IF) and Western blot. We found that butyrate activated the histone H3 acetylation of MG-63 cells. Exposure of MG-63 cells to butyrate partly decreased cell viability with no marked increase in apoptosis and necrosis. Twenty-four hours of exposure to butyrate stimulated RANKL protein expression, whereas it inhibited OPG protein expression. Butyrate also inhibited the secretion of OPG in MG-63 cells, whereas the sRANKL level was below the detection limit. However, 3 days of exposure to butyrate (1 to 8 mM) or other HDAC inhibitors such as phenylbutyrate, valproic acid and trichostatin stimulated OPG secretion. Butyrate stimulated 8-isoprostane, MMP-2 and OPN secretion, but not procollagen I, or osteocalcin in MG-63 cells. Exposure to butyrate (2⁻4 mM) for 3 days markedly stimulated osteonectin secretion and ALP activity. In conclusion, higher concentrations of butyric acid generated by periodontal and root canal microorganisms may potentially induce bone destruction and impair bone repair by the alteration of OPG/RANKL expression/secretion, 8-isoprostane, MMP-2 and OPN secretion, and affect cell viability. However, lower concentrations of butyrate (1⁻4 mM) may stimulate ALP, osteonectin and OPG. These effects are possibly related to increased histone acetylation. These events are important in the pathogenesis and repair of periodontal and periapical destruction.


Assuntos
Butiratos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Histonas/metabolismo , Isoprostanos/biossíntese , Osteoblastos/metabolismo , Osteoprotegerina/biossíntese , Ligante RANK/biossíntese , Acetilação/efeitos dos fármacos , Butiratos/metabolismo , Linhagem Celular , Cavidade Pulpar/metabolismo , Cavidade Pulpar/microbiologia , Cavidade Pulpar/patologia , Histonas/genética , Humanos , Isoprostanos/genética , Osteoblastos/patologia , Osteoprotegerina/genética , Periodontite/genética , Periodontite/metabolismo , Periodontite/microbiologia , Periodontite/patologia , Ligante RANK/genética
13.
Molecules ; 23(9)2018 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-30235836

RESUMO

Postmenopausal diabetic women have a high risk of fractures. Salidroside has preventive effects on estrogen deficiency-induced osteoporosis and has hypoglycemic effects on diabetes in rats. However, whether salidroside inhibits bone loss in postmenopausal diabetic patients is still unknown. Here, we established a rat model of osteoporosis to investigate the protective effects of salidroside on bone loss induced by ovariectomy combined with diabetes, also investigating the underlying mechanisms. Two-month-old female Sprague-Dawley rats were divided into three equal groups (10 rats in each group): control group (with sham operation, treated with drug vehicle); OVX/T1DM group (ovariectomized diabetic rats); OVX/T1DM-SAL group, comprising ovariectomized diabetic rats treated with salidroside (20 mg/kg body weight) by gavage. The results showed that after 60 consecutive days of treatment, the bone mineral density (BMD) of OVX/T1DM-SAL increased significantly compared with the OVX/T1DM group (p < 0.01). The level of serum bone turnover markers, including alkaline phosphatase (ALP), cross linked c-telopeptide of type I collagen (CTX-1), osteocalcin, N-terminal propeptide of type I procollagen (PINP), and tartrate-resistant acid phosphatase 5b (TRACP 5b) were all increased in the OVX/T1DM group compared with the control (p < 0.01), and those were decreased by salidroside treatment. Meanwhile, the bone histopathological changes were also attenuated, and the bone marrow adipogenesis was inhibited in salidroside treated rats. Moreover, protein and mRNA ratio of bone osteoprotegerin (OPG)/receptor activator of nuclear factor-κB ligand (RANKL) was upregulated in ovariectomized diabetic rats by salidroside treatment. The results above indicated that the protective effect of salidroside on bone loss induced by ovariectomy and diabetes was mainly due to its ability to suppress bone turnover, inhibit bone marrow adipogenesis, and up-regulate the OPG/RANKL ratio.


Assuntos
Conservadores da Densidade Óssea/uso terapêutico , Reabsorção Óssea/prevenção & controle , Diabetes Mellitus/patologia , Glucosídeos/uso terapêutico , Osteoporose/prevenção & controle , Osteoprotegerina/biossíntese , Fenóis/uso terapêutico , Ligante RANK/biossíntese , Adipogenia/efeitos dos fármacos , Animais , Densidade Óssea/efeitos dos fármacos , Reabsorção Óssea/tratamento farmacológico , Osso e Ossos/metabolismo , Modelos Animais de Doenças , Feminino , Osteoporose/tratamento farmacológico , Ovariectomia , Ratos , Ratos Sprague-Dawley
14.
Eur J Orthod ; 40(4): 356-363, 2018 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-29016746

RESUMO

Objective: Vibration can be used to accelerate tooth movement, though the exact mechanisms remain unclear. This study aimed to investigate the effects of low magnitude high frequency (LMHF) vibration combined with compressive force on periodontal ligament (PDL) cells in vitro. Materials and methods: Human PDL cells were isolated from extracted premolar teeth of four individuals. To determine the optimal frequency for later used in combination with compressive force, three cycles of low-magnitude (0.3 g) vibrations at various frequencies (30, 60, or 90 Hz) were applied to PDL cells for 20 min every 24 h. To investigate the effects of vibration combined with compressive force, PDL cells were subjected to three cycles of optimal vibration frequency (V) or 1.5 g/cm2 compressive force for 48 h (C) or vibration combined with compressive force (VC). Cell viability was assessed using MTT assay. PGE2, soluble RANKL (sRANKL), and OPG production were quantified by ELISA. RANKL, OPG, and Runx2 expression were determined using real-time PCR. Results: Cell viability was decreased in groups C and VC. PGE2 and RANKL, but not OPG, were increased in groups V, C, and VC, thus increasing the RANKL/OPG ratio. The highest level was observed in group VC. sRANKL was increased in groups V, C, and VC; however, no significant different between the experimental groups. Runx2 expression was reduced in groups C and VC. Conclusions: Vibration increased PGE2, RANKL, and sRANKL, but not OPG and Runx2. Vibration had the additive effects on PGE2 and RANKL, but not sRANKL in compressed PDL cells.


Assuntos
Ligamento Periodontal/citologia , Vibração , Sobrevivência Celular , Células Cultivadas , Dinoprostona/biossíntese , Ensaio de Imunoadsorção Enzimática , Humanos , Mecanotransdução Celular/fisiologia , Osteoprotegerina/biossíntese , Ligamento Periodontal/metabolismo , Ligante RANK/biossíntese , Reação em Cadeia da Polimerase em Tempo Real , Estresse Mecânico , Técnicas de Movimentação Dentária
15.
J BUON ; 23(4): 1029-1040, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30358208

RESUMO

PURPOSE: RANKL, OPG and TRAIL have long been pursued in cancer. Mutated KRas proteins and c-Fos overexpression - well-recognized oncogenic events - have been conceived as coordinators of RANKL, OPG and TRAIL pathways. Considering the paucity in the relevant literature, the purpose of the present study was to investigate whether the expression of these molecules configures a distinct papillary thyroid carcinoma (PTC) subgroup with adverse clinicopathological characteristics. METHODS: RANKL, OPG, TRAIL, KRas, and c-Fos immunohistochemical expression in relation to clinicopathological characteristics of PTC was assessed retrospectively in paraffin-embedded PTC specimens from 114 patients who underwent total thyroidectomy with simultaneous central lymph node dissection (CLND). RESULTS: Expression of RANKL, OPG, TRAIL, Kras and c- Fos was revealed in 78.6, 63.2, 61.4, 47.4, and 73.7% of PTC, respectively. As predominant KRas-expressing PTC histotype emerged the classical PTC (cPTC), comprising 66.7% of PTC. A significant correlation was demonstrated of RANKL, OPG, and TRAIL expression with central lymph node metastasis CLNM (p=0.007, p<0.001, and p=0.002, respectively), concerning especially cPTC as regards to RANKL (p=0.027) and OPG (p=0.006), and both cPTC (p=0.043) and follicular variant of PTC (FVPTC) (p=0.049) with regard to TRAIL. OPG expression associated significantly with multifocality (p=0.045). Multivariable-adjusted logistic regression models characterized TRAIL as independent predictor of CLNM (OR=10.335, 95% CI: 1.23-86.87). CLNM correlated significantly with six pairs of coexpressions: TRAIL-KRas (p=0.011), TRAIL-c-Fos (p=0.006), OPG-c-Fos (p=0.024), RANKL-TRAIL (p<0.001), RANKL-OPG (p<0.001), TRAIL- OPG (p<0.001). CONCLUSION: The present study suggested for the first time that OPG, RANKL, TRAIL expressions, either alone or in concert involving c-Fos and KRas expression, are related to CLNM. Further research is warranted to elucidate whether the examined molecules can be endorsed as indicators of aggressive PTC behavior and guide a personalized therapeutic intervention.


Assuntos
Câncer Papilífero da Tireoide/metabolismo , Câncer Papilífero da Tireoide/patologia , Neoplasias da Glândula Tireoide/metabolismo , Adulto , Feminino , Humanos , Imuno-Histoquímica , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Oncogenes , Osteoprotegerina/biossíntese , Proteínas Proto-Oncogênicas c-fos/biossíntese , Proteínas Proto-Oncogênicas p21(ras)/biossíntese , Proteínas Proto-Oncogênicas p21(ras)/genética , Ligante RANK/biossíntese , Estudos Retrospectivos , Ligante Indutor de Apoptose Relacionado a TNF/biossíntese , Câncer Papilífero da Tireoide/genética , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/patologia
16.
J Biol Chem ; 291(48): 24912-24921, 2016 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-27729453

RESUMO

Chondrogenesis can regulate bone formation. Fibroblast growth factor receptor 3, highly expressed in chondrocytes, is a negative regulator of bone growth. To investigate whether chondrocyte FGFR3 regulates osteogenesis, thereby contributing to postnatal bone formation and bone remodeling, mice with conditional knock-out of Fgfr3 in chondrocytes (mutant (MUT)) were generated. MUT mice displayed overgrowth of bone with lengthened growth plates. Bone mass of MUT mice was significantly increased at both 1 month and 4 months of age. Histological analysis showed that osteoblast number and bone formation were remarkably enhanced after deletion of Fgfr3 in chondrocytes. Chondrocyte-osteoblast co-culture assay further revealed that Fgfr3 deficiency in chondrocytes promoted differentiation and mineralization of osteoblasts by up-regulating the expressions of Ihh, Bmp2, Bmp4, Bmp7, Wnt4, and Tgf-ß1, as well as down-regulating Nog expression. In addition, osteoclastogenesis was also impaired in MUT mice with decreased number of osteoclasts lining trabecular bone, which may be related to the reduced ratio of Rankl to Opg in Fgfr3-deficient chondrocytes. This study reveals that chondrocyte FGFR3 is involved in the regulation of bone formation and bone remodeling by a paracrine mechanism.


Assuntos
Condrócitos/metabolismo , Lâmina de Crescimento/metabolismo , Osteogênese/fisiologia , Osteoprotegerina/biossíntese , Comunicação Parácrina/fisiologia , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/biossíntese , Animais , Proteínas Morfogenéticas Ósseas/biossíntese , Proteínas Morfogenéticas Ósseas/genética , Remodelação Óssea/fisiologia , Células Cultivadas , Condrócitos/citologia , Técnicas de Cocultura , Lâmina de Crescimento/citologia , Camundongos , Camundongos Knockout , Tamanho do Órgão/fisiologia , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteoclastos/citologia , Osteoclastos/metabolismo , Osteoprotegerina/genética , Ligante RANK/biossíntese , Ligante RANK/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Fator de Crescimento Transformador beta1/genética , Proteína Wnt4/biossíntese , Proteína Wnt4/genética
17.
Biochim Biophys Acta ; 1860(10): 2211-9, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27154288

RESUMO

BACKGROUND: The balance between osteoblastic and osteoclastic activity is critical in orthodontic tooth movement (OTM). Mesenchymal stem cells (MSCs) play an important role in maintaining bone homeostasis, and periodontal ligament stem cells (PDLSCs) are tissue-specific MSCs in the periodontal ligament. However, whether PDLSCs are required for periodontal tissue remodeling during OTM is not fully understood. METHODS: Here, we used PDGFRα and Nestin to trace PDLSCs during OTM in rats. We treat human PDLSCs with 100kpa static pressure for 1h or 12h in vitro, and examined the phenotypic changes and expression of RANKL and OPG in these cells. RESULTS: In vivo, we found that positive signals of PDGFRα and Nestin in the PDL gradually increased and then decreased on the pressure side to which pressure was applied. In vitro, the osteogenic differentiation of PDLSCs was significantly increased after force treatment for 1h relative to 12h. In contrast, the expression ratio of RANKL/OPG was reduced at 1h and significantly increased at 12h. Furthermore, we found that the Wnt/ß-catenin pathway was dynamically activated in the PDL and in PDLSCs after mechanical stimulation. Importantly, the canonical Wnt pathway inhibitor DKK1 blocked the osteogenesis effect and rescued the ratio of RANKL/OPG in PDLSCs under force treatment for 1h. CONCLUSIONS: Our findings reveal that PDLSCs participate in OTM and that the Wnt/ß-catenin pathway maintains bone homeostasis during tooth movement by regulating the balance between osteoblastic and osteoclastic activity. GENERAL SIGNIFICANCE: We describe a novel potential mechanism related to tooth movement.


Assuntos
Diferenciação Celular/genética , Osteogênese/genética , Osteoprotegerina/biossíntese , Ligante RANK/biossíntese , Estresse Mecânico , Animais , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Células-Tronco Mesenquimais/metabolismo , Nestina/genética , Nestina/metabolismo , Osteoprotegerina/genética , Ligamento Periodontal/crescimento & desenvolvimento , Ligamento Periodontal/metabolismo , Ligante RANK/genética , Ratos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Técnicas de Movimentação Dentária , Via de Sinalização Wnt/genética
18.
Prostaglandins Other Lipid Mediat ; 128-129: 27-33, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28163121

RESUMO

(-)-Epigallocatechin gallate (EGCG), the most abundant flavonoid in green tea, and chlorogenic acid, the main polyphenol found in coffee, attract significant attention owing to health benefits. We have previously demonstrated that prostaglandin E2 (PGE2) stimulates osteoprotegerin synthesis through the activation of p38 mitogen-activated protein (MAP) kinase and stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) in osteoblast-like MC3T3-E1 cells. In the present study, we investigated the effects of EGCG or chlorogenic acid on the PGE2-stimulated osteoprotegerin synthesis in MC3T3-E1 cells. EGCG significantly amplified the PGE2-induced release. EGCG markedly enhanced the expression levels of osteoprotegerin mRNA induced by PGE2. On the contrary, chlorogenic acid had no effect on the PGE2-stimulated release of osteoprotegerin. EGCG significantly strengthened the PGE2-induced phosphorylation of p38 MAP kinase and SAPK/JNK, whereas chlorogenic acid failed to affect them. BIRB0796 and SP600125, a p38 MAP kinase inhibitor and a SAPK/JNK inhibitor, respectively, markedly reduced the amplification by EGCG of the PGE2-stimulated osteoprotegerin release. These results strongly suggest that EGCG synergistically enhances the PGE2-stimulated osteoprotegerin synthesis via potentiation of p38 MAP kinase and SAPK/JNK in osteoblasts. Our present findings could present a new significant aspect in the favorable effect of EGCG on the prevention of osteoporotic bone loss and fracture especially in elderly people since osteoprotegerin secreted from osteoblasts is well-recognized to act as a suppressor of osteoclastic bone resorption.


Assuntos
Catequina/análogos & derivados , Dinoprostona/farmacologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoprotegerina/biossíntese , Animais , Catequina/farmacologia , Linhagem Celular , Sinergismo Farmacológico , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Osteoprotegerina/genética , Fosforilação/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Regulação para Cima/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
J Immunol ; 195(6): 2675-82, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26254339

RESUMO

In the thymus, medullary thymic epithelial cells (mTEC) regulate T cell tolerance via negative selection and Foxp3(+) regulatory T cell (Treg) development, and alterations in the mTEC compartment can lead to tolerance breakdown and autoimmunity. Both the receptor activator for NF-κB (RANK)/RANK ligand (RANKL)/osteoprotegerin (OPG) axis and expression of the transcriptional regulator Aire are involved in the regulation of thymus medullary microenvironments. However, their impact on the mechanisms controlling mTEC homeostasis is poorly understood, as are the processes that enable the thymus medulla to support the balanced production of mTEC-dependent Foxp3(+) Treg. In this study, we have investigated the control of mTEC homeostasis and examined how this process impacts the efficacy of Foxp3(+) Treg development. Using newly generated RANK Venus reporter mice, we identify distinct RANK(+) subsets that reside within both the mTEC(hi) and mTEC(lo) compartments and that represent direct targets of OPG-mediated control. Moreover, by mapping OPG expression to a subset of Aire(+) mTEC, our data show how cis- and trans-acting mechanisms are able to control the thymus medulla by operating on multiple mTEC targets. Finally, we show that whereas the increase in mTEC availability in OPG-deficient (Tnfrsf11b(-/-)) mice impacts the intrathymic Foxp3(+) Treg pool by enhancing peripheral Treg recirculation back to the thymus, it does not alter the number of de novo Rag2pGFP(+)Foxp3(+) Treg that are generated. Collectively, our study defines patterns of RANK expression within the thymus medulla, and it shows that mTEC homeostasis is not a rate-limiting step in intrathymic Foxp3(+) Treg production.


Assuntos
Linfopoese/imunologia , Osteoprotegerina/genética , Ligante RANK/imunologia , Linfócitos T Reguladores/imunologia , Timo/metabolismo , Animais , Autoimunidade/imunologia , Células Cultivadas , Proteínas de Ligação a DNA/genética , Células Epiteliais , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Tolerância Imunológica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/imunologia , Técnicas de Cultura de Órgãos , Osteoprotegerina/biossíntese , Osteoprotegerina/imunologia , Ligante RANK/biossíntese , Transdução de Sinais/imunologia , Linfócitos T Reguladores/citologia , Timo/citologia , Timo/imunologia , Fatores de Transcrição/biossíntese , Proteína AIRE
20.
Appl Microbiol Biotechnol ; 101(12): 4923-4933, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28303296

RESUMO

As a natural inhibitor of the receptor activator of nuclear factor-кB ligand (RANKL), osteprotegerin (OPG) is considered a promising treatment for metabolic bone diseases. Typical approaches for preparing recombinant OPG or its derivatives employ eukaryotic expression systems. Due to the advantages of a prokaryotic expression system, which include its convenience, low cost, and abundant production, in this study, we establish a strategy for preparing functional OPG using the Escherichia coli expression system. After initial failures in preparation of OPG and its truncation, OPG cysteine-rich domain (OPG-CRD/OPGT) by using pET and pGEX vectors, we constructed a sortase A (SrtA)-aided E. coli expression system, in which the expressed protein was a self-cleaving SrtA fusion protein. Using this system, we successfully prepared the recombinant OPGT protein. The BIAcore analyses indicated that the prepared OPGT had high affinities in binding with RANKL and TRAIL. Cell experiments confirmed the inhibitory effects of the prepared OPGT on RANKL-induced osteoclast differentiation and TRAIL-induced tumor cell apoptosis. The sortase A-aided E. coli expression system for OPGT established in this study may contribute to further studies and commercial applications of OPG.


Assuntos
Aminoaciltransferases/metabolismo , Proteínas de Bactérias/metabolismo , Cisteína Endopeptidases/metabolismo , Cisteína/química , Escherichia coli/genética , Osteoprotegerina/química , Osteoprotegerina/genética , Aminoaciltransferases/genética , Animais , Apoptose/efeitos dos fármacos , Proteínas de Bactérias/genética , Diferenciação Celular/efeitos dos fármacos , Cisteína/genética , Cisteína Endopeptidases/genética , Escherichia coli/enzimologia , Vetores Genéticos , Humanos , Camundongos , Osteoclastos/efeitos dos fármacos , Osteoprotegerina/biossíntese , Osteoprotegerina/farmacologia , Ligação Proteica , Domínios Proteicos , Ligante RANK/farmacologia , Células RAW 264.7 , Proteínas Recombinantes/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa