Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int Immunopharmacol ; 101(Pt A): 108177, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34626872

RESUMO

Osteoporosis, characterized by bone loss and microstructure damage, occurs when osteoclast activity outstrips osteoblast activity. Natural compounds with inhibitory effect on osteoclast differentiation and function have been evidenced to protect from osteoporosis. After multiple compounds screening, 12-deoxyphorbol 13-acetate (DPA) was found to decline RANKL-induced osteoclastogenesis dose-dependently by attenuating activities of NFATc1 and c-Fos, followed by decreasing the level of osteoclast function-associated genes and proteins including Acp5, V-ATPase-d2 and CTSK. Mechanistically, we found that DPA suppressing RANKL-induced downstream signaling pathways, including MAPK signaling pathway and calcium oscillations. Furthermore, the in vivo efficacy of DPA was further confirmed in an OVX-induced osteoporosis mice model. Collectively, the results in our presentation reveal that DPA might be a promising compound to manage osteoporosis.


Assuntos
Fatores de Transcrição NFATC/antagonistas & inibidores , Osteoporose/tratamento farmacológico , Ésteres de Forbol/farmacologia , Animais , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/imunologia , Camundongos , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoclastos/fisiologia , Osteogênese/efeitos dos fármacos , Osteogênese/imunologia , Osteoporose/imunologia , Ésteres de Forbol/uso terapêutico , Células RAW 264.7
2.
Int Immunopharmacol ; 98: 107869, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34153673

RESUMO

OBJECTIVE: Spondyloarthritis (SpA) is mainly characterized by bone erosion, new bone formation, inflammation and potential disability. Epigallocatechin gallate (EGCG) has been proved to be closely related with the regulation of inflammation and bone metabolism. However, whether EGCG could improve SpA remains unclear. METHODS: SpA animal model was established using proteoglycan. Cell proliferation were measured by CCK-8 assay. The mRNA expression levels of genes were detected using qRT-PCR, protein levels were assessed via western blotting and immunohistochemistry. ELISA assay was performed to examined the inflammatory cytokine release. Lesions in spine cartilage tissues were observed using hematoxylin-eosin (HE) and Safranin O staining. Alkaline phosphatase (ALP) assay and Alizarin Red S staining was used to investigate osteoblast mineralization. RESULTS: We found that EGCG could inhibit inflammation and new bone formation in SpA mice. Besides, inflammatory factor expression and osteogenic differentiation in osteoblasts isolated from SpA mice were also decreased by EGCG. Further, EGCG treatment suppressed the activation of Wnt/ß-Catenin/COX-2 pathway and the activator of this pathway partially reversed the effects of EGCG on inflammation and osteoblast differentiation. CONCLUSIONS: EGCG repressed inflammatory responses and new bone formation, and further improved SpA through Wnt/ß-Catenin/COX-2 pathway. Our findings may provide a new thought for the prevention and treatment of SpA.


Assuntos
Artrite Experimental/tratamento farmacológico , Catequina/análogos & derivados , Osteogênese/efeitos dos fármacos , Espondilartrite/tratamento farmacológico , Via de Sinalização Wnt/efeitos dos fármacos , Administração Oral , Animais , Artrite Experimental/imunologia , Catequina/farmacologia , Catequina/uso terapêutico , Ciclo-Oxigenase 2/metabolismo , Avaliação Pré-Clínica de Medicamentos , Humanos , Masculino , Camundongos , Osteogênese/imunologia , Espondilartrite/imunologia , Via de Sinalização Wnt/imunologia , beta Catenina/metabolismo
3.
Int J Mol Sci ; 21(12)2020 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-32630456

RESUMO

Jellyfish collagen, which can be defined as "collagen type 0" due to its homogeneity to the mammalian types I, II, III, V, and IX and its batch-to-batch consistent producibility, is of special interest for different medical applications related to (bone) tissue regeneration as an alternative to mammalian collagen-based biomaterials. However, no in vivo studies regarding the induction of M1- and M2-macrophages and their time-dependent ration as well as the analysis of the bone regeneration capacity of jellyfish collagen scaffolds have been conducted until now. Thus, the goal of this study was to determine the nature of the immune response to jellyfish collagen scaffolds and their bone healing capacities. Two in vivo studies using established implantation models, i.e., the subcutaneous and the calvarian implantation model in Wistar rats, were conducted. Furthermore, specialized histological, histopathological, and histomorphometrical methods have been used. As a control biomaterial, a collagen scaffold, originating from porcine pericardium, which has already been stated as biocompatible, was used for the subcutaneous study. The results of the present study show that jellyfish collagen scaffolds are nearly completely resorbed until day 60 post implantation by stepwise integration within the subcutaneous connective tissue mediated mainly by macrophages and single multinucleated giant cells. Interestingly, the degradation process ended in a vessel rich connective tissue that is understood to be an optimal basis for tissue regeneration. The study results showed an overall weaker immune response to jellyfish collagen than to porcine pericardium matrices by the induction of significantly lower numbers of macrophages together with a more balanced occurrence of M1- and M2-macrophages. However, both collagen-based biomaterials induced balanced numbers of both macrophage subtypes, which supports their good biocompatibility. Moreover, the histomorphometrical results for the calvarial implantation of the jellyfish scaffolds revealed an average of 46.20% de novo bone formation at day 60, which was significantly higher compared to the control group. Thereby, the jellyfish collagen scaffolds induced also significantly higher numbers of anti-inflammatory macrophages within the bony implantation beds. Altogether, the results show that the jellyfish collagen scaffolds allowed for a directed integration behavior, which is assumed to be in accordance with the concept of Guided Bone Regeneration (GBR). Furthermore, the jellyfish collagen scaffolds induced a long-term anti-inflammatory macrophage response and an optimal vascularization pattern within their implant beds, thus showing excellent biocompatibility and (bone) tissue healing properties.


Assuntos
Regeneração Óssea/fisiologia , Colágeno/metabolismo , Engenharia Tecidual/métodos , Animais , Materiais Biocompatíveis/metabolismo , Regeneração Óssea/genética , Osso e Ossos/imunologia , Osso e Ossos/metabolismo , Colágeno/imunologia , Imunidade , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Osteogênese/imunologia , Osteogênese/fisiologia , Ratos , Ratos Wistar , Cifozoários/metabolismo , Alicerces Teciduais , Cicatrização/fisiologia
4.
Korean J Intern Med ; 34(1): 210-219, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-28286938

RESUMO

BACKGROUND/AIMS: This study aimed to determine the regulatory role of N-acetyl-l-cysteine (NAC), an antioxidant, in interleukin 17 (IL-17)-induced osteoclast differentiation in rheumatoid arthritis (RA). METHODS: After RA synovial fibroblasts were stimulated by IL-17, the expression and production of receptor activator of nuclear factor κ-B ligand (RANKL) was determined by real-time polymerase chain reaction and enzyme-linked immunosorbent assay (ELISA). Osteoclastogenesis was also determined after co-cultures of IL-17-stimulated RA synovial fibroblasts, Th17 cells and various concentrations of NAC with monocytes. After human peripheral CD4+ T cells were cultured with NAC under Th17 condition, IL-17, interferon γ, IL-4, Foxp3, RANKL, and IL-2 expression and production was determined by flow cytometry or ELISA. RESULTS: When RA synovial fibroblasts were stimulated by IL-17, IL-17 stimulated the production of RANKL, and NAC reduced the IL-17-induced RANKL production in a dose-dependent manner. NAC decreased IL-17-activated phosphorylation of mammalian target of rapamycin, c-Jun N-terminal kinase, and inhibitor of κB. When human peripheral blood CD14+ monocytes were cultured with macrophage colony-stimulating factor and IL-17 or RANKL, osteoclasts were differentiated, and NAC reduced the osteoclastogenesis. After human peripheral CD4+ T cells were co-cultured with IL-17-pretreated RA synovial fibroblasts or Th17 cells, NAC reduced their osteoclastogenesis. Under Th17 polarizing condition, NAC decreased Th17 cell differentiation and IL-17 and RANKL production. CONCLUSION: NAC inhibits the IL-17-induced RANKL production in RA synovial fibroblasts and IL-17-induced osteoclast differentiation. NAC also reduced Th17 polarization. NAC could be a supplementary therapeutic option for inflammatory and bony destructive processes in RA.


Assuntos
Acetilcisteína/farmacologia , Artrite Reumatoide/tratamento farmacológico , Osteogênese/efeitos dos fármacos , Ligante RANK/metabolismo , Células Th17/efeitos dos fármacos , Adulto , Idoso , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/imunologia , Fibroblastos/patologia , Humanos , Técnicas In Vitro , Interleucina-17/metabolismo , Pessoa de Meia-Idade , Osteogênese/imunologia , Ligante RANK/genética , Transdução de Sinais/efeitos dos fármacos , Membrana Sinovial/efeitos dos fármacos , Membrana Sinovial/imunologia , Membrana Sinovial/patologia , Células Th17/imunologia , Células Th17/patologia
5.
Arthritis Rheumatol ; 68(12): 2889-2900, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27563728

RESUMO

OBJECTIVE: Proinflammatory molecules promote osteoclast-mediated bone erosion by up-regulating local RANKL production. However, recent evidence suggests that combinations of cytokines, such as tumor necrosis factor (TNF) plus interleukin-6 (IL-6), induce RANKL-independent osteoclastogenesis. The purpose of this study was to better understand TNF/IL-6-induced osteoclast formation and to determine whether RANK is absolutely required for osteoclastogenesis and bone erosion in murine inflammatory arthritis. METHODS: Myeloid precursors from wild-type (WT) mice or mice with either germline or conditional deletion of Rank, Nfatc1, Dap12, or Fcrg were treated with either RANKL or TNF plus IL-6. Osteoprotegerin, anti-IL-6 receptor (anti-IL-6R), and hydroxyurea were used to block RANKL, the IL-6R, and cell proliferation, respectively. Clinical scoring, histologic assessment, micro-computed tomography, and quantitative polymerase chain reaction (qPCR) were used to evaluate K/BxN serum-transfer arthritis in WT and RANK-deleted mice. Loss of Rank was verified by qPCR and by osteoclast cultures. RESULTS: TNF/IL-6 generated osteoclasts in vitro that resorbed mineralized tissue through a pathway dependent on IL-6R, NFATc1, DNAX-activation protein 12, and cell proliferation, but independent of RANKL or RANK. Bone erosion and osteoclast formation were reduced, but not absent, in arthritic mice with inducible deficiency of RANK. TNF/IL-6, but not RANKL, induced osteoclast formation in bone marrow and synovial cultures from animals deficient in Rank. Multiple IL-6 family members (IL-6, leukemia inhibitory factor, oncostatin M) were up-regulated in the synovium of arthritic mice. CONCLUSION: The persistence of bone erosion and synovial osteoclasts in Rank-deficient mice, and the ability of TNF/IL-6 to induce osteoclastogenesis, suggest that more than one cytokine pathway exists to generate these bone-resorbing cells in inflamed joints.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Artrite Experimental/genética , Reabsorção Óssea/genética , Fatores de Transcrição NFATC/genética , Osteogênese/genética , Receptor Ativador de Fator Nuclear kappa-B/genética , Animais , Artrite Experimental/imunologia , Reabsorção Óssea/imunologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Inibidores Enzimáticos/farmacologia , Hidroxiureia/farmacologia , Técnicas In Vitro , Interleucina-6/farmacologia , Camundongos , Camundongos Knockout , Osteogênese/efeitos dos fármacos , Osteogênese/imunologia , Osteoprotegerina/farmacologia , Ligante RANK/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Receptores de IgG/genética , Receptores de Interleucina-6/antagonistas & inibidores , Fator de Necrose Tumoral alfa/farmacologia , Microtomografia por Raio-X
6.
J Biomater Sci Polym Ed ; 24(15): 1794-813, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23746285

RESUMO

Recent insight into the critical role of pro-inflammatory cytokines, particularly tumor necrosis factor-α (TNF-α), in bone regeneration has heralded a new direction in the design of tissue engineering constructs. Previous studies have demonstrated that continuous delivery of 50 ng/ml TNF-α to mesenchymal stem cells (MSCs) cultured on three-dimensional (3D) biodegradable electrospun poly(ϵ-caprolactone) (PCL) microfiber meshes stimulates mineralized matrix deposition, a marker of osteogenic differentiation. Since TNF-α exhibits a biphasic pattern of expression following bone fracture in vivo, this study aimed to investigate the effects of temporal patterns of TNF-α delivery on in vitro osteogenic differentiation of MSCs cultured on 3D electrospun PCL scaffolds. MSCs were cultured for 16 days and exposed to continuous, early, intermediate, or late TNF-α delivery. To further elucidate the effects of TNF-α on osteogenic differentiation, the study design included MSCs precultured both in the presence and absence of typically required osteogenic supplement dexamethasone. Mineralized matrix deposition was not observed in constructs with dexamethasone-naïve MSCs, suggesting that TNF-α is not sufficient to trigger in vitro osteogenic differentiation of MSCs. For MSCs precultured with dexamethasone, TNF-α suppressed alkaline phosphatase activity, an early marker of osteogenic differentiation, and stimulated mineralized matrix deposition, a late stage marker of MSC osteogenic differentiation. By elucidating the impact of temporal variations in TNF-α delivery on MSC osteogenic differentiation, our results offer insight into the regenerative mechanism of TNF-α and provide the design parameters for a novel tissue engineering strategy that rationally controls TNF-α signaling to stimulate bone regeneration.


Assuntos
Células-Tronco Mesenquimais/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Poliésteres/química , Alicerces Teciduais/química , Fator de Necrose Tumoral alfa/administração & dosagem , Animais , Regeneração Óssea/efeitos dos fármacos , Regeneração Óssea/imunologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Histocitoquímica , Masculino , Células-Tronco Mesenquimais/citologia , Microscopia Eletrônica de Varredura , Osteogênese/imunologia , Ratos , Ratos Endogâmicos F344 , Engenharia Tecidual/métodos , Fator de Necrose Tumoral alfa/imunologia
7.
Osteoarthritis Cartilage ; 10(12): 968-76, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12464557

RESUMO

OBJECTIVE: Vitamin A derivatives are widely used therapeutic agents for the treatment of dermatological and rheumatological disorders. Long-standing administration of these drugs, in turn, causes skeletal changes including ossification of ligaments, premature fusion of epiphyses and abnormalities of modeling. Recent in vitro experiments have further suggested that retinoid treatment of cultured chondrocytes may cause apoptotic cell death. The present study aims to address detailed cartilage changes associated with in vivo administration of vitamin A derivatives. METHODS: Retinyl acetate was administrated to experimental mice, C3H-Heston, for more than 12 months. Modified morphometry on the articular cartilage and fluorescent labeling of the subchondral bone were carried out to address the changes in the articular cartilage and subchondral bone. In order to address the detailed chondrocytes phenotypes, electron microscopy was carried out. Since findings of these studies suggested that biological properties of the cartilage matrix might be altered, the present study also immunolocalized functional matrix molecules, type I collagen and osteoblast-stimulating factor-1 (OSF-1). RESULTS: Histomorphometry demonstrated that retinoid administration lead to progressive atrophy of the articular cartilage with concomitant proliferation of subchondral bone. Furthermore, detailed light and electron microscopy suggested that the subchondral bone proliferates into the degenerating cartilage. The affected articular cartilage also resembled that of osteoarthritis in terms of ectopic type I collagen production. Furthermore, the affected articular cartilage produced a developmentally regulated matrix molecule, osteoblast-stimulating factor-1 (OSF-1) that is normally expressed in both the fetal cartilage and the epiphyseal growth plate cartilage but not in the articular cartilage. CONCLUSION: The present results indicate that the systemic retinoid administration may alter the biological properties of the articular cartilage.


Assuntos
Adjuvantes Imunológicos/farmacologia , Cartilagem Articular/efeitos dos fármacos , Condrócitos/metabolismo , Vitamina A/análogos & derivados , Vitamina A/farmacologia , Animais , Atrofia/imunologia , Atrofia/fisiopatologia , Cartilagem Articular/patologia , Cartilagem Articular/fisiopatologia , Diferenciação Celular , Condrócitos/efeitos dos fármacos , Condrócitos/fisiologia , Diterpenos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Microscopia Eletrônica , Osteoblastos/imunologia , Osteoblastos/fisiologia , Osteogênese/imunologia , Osteogênese/fisiologia , Fenótipo , Ésteres de Retinil
8.
J Immunol ; 169(5): 2374-80, 2002 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12193704

RESUMO

We examined the ability of 1,25 (OH)(2) vitamin D(3) (Vit D) to stimulate osteoclast-like cell (OCL) formation in cocultures of spleen cells and primary calvarial osteoblasts from wild-type (WT) and IL-1R type 1-deficient (knockout; KO) mice. Vit D dose dependently increased OCL in cocultures containing WT osteoblasts. In contrast, there was a 90% reduction in OCL numbers in cocultures containing KO osteoblasts. In cocultures with either WT or KO osteoblasts, treatment with Vit D increased receptor activator of NF-kappaB ligand mRNA by 17-, 19-, or 3.5-fold, respectively. Vit D decreased osteoprotegerin mRNA to undetectable in all groups. Intracellular IL-1alpha protein increased after Vit D treatment in cocultures containing WT, but not KO osteoblasts. We also examined direct effects of Vit D, IL-1alpha, and their combination on gene expression in primary osteoblasts. In WT cells, Vit D and IL-1 stimulated receptor activator of NF-kappaB ligand mRNA expression by 3- and 4-fold, respectively, and their combination produced a 7-fold increase. Inhibition of osteoprotegerin mRNA in WT cells was partial with either agent alone and greatest with their combination. In KO cells, only Vit D stimulated a response. IL-1 alone increased IL-1alpha protein expression in WT osteoblasts. However, in combination with Vit D, there was a synergistic response (100-fold increase). In KO cultures, there were no effects of IL-1, Vit D, or their combination on IL-1alpha protein. These results demonstrate interactions between IL-1 and Vit D in primary osteoblasts that appear important in both regulation of IL-1alpha production and the ability of Vit D to support osteoclastogenesis.


Assuntos
Calcitriol/farmacologia , Proteínas de Transporte/biossíntese , Glicoproteínas/metabolismo , Interleucina-1/biossíntese , Glicoproteínas de Membrana/biossíntese , Osteoblastos/metabolismo , Osteoclastos/citologia , Osteogênese/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/metabolismo , Baço/citologia , Adjuvantes Imunológicos/biossíntese , Adjuvantes Imunológicos/fisiologia , Animais , Calcitriol/antagonistas & inibidores , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Células Cultivadas , Técnicas de Cocultura , Glicoproteínas/antagonistas & inibidores , Glicoproteínas/genética , Proteína Antagonista do Receptor de Interleucina 1 , Interleucina-1/antagonistas & inibidores , Interleucina-1/genética , Interleucina-1/fisiologia , Ligantes , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Osteoblastos/efeitos dos fármacos , Osteoblastos/fisiologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/fisiologia , Osteogênese/imunologia , Osteoprotegerina , Ligante RANK , RNA Mensageiro/biossíntese , Receptor Ativador de Fator Nuclear kappa-B , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/genética , Receptores do Fator de Necrose Tumoral , Sialoglicoproteínas/farmacologia , Baço/efeitos dos fármacos , Baço/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA