Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 21(24)2020 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-33371439

RESUMO

Transforming growth factor ß1 (TGFß1) is a major mediator in the modulation of osteoblast differentiation. However, the underlying molecular mechanism is still not fully understood. Here, we show that TGFß1 has a dual stage-dependent role in osteoblast differentiation; TGFß1 induced matrix maturation but inhibited matrix mineralization. We discovered the underlying mechanism of the TGFß1 inhibitory role in mineralization using human osteoprogenitors. In particular, the matrix mineralization-related genes of osteoblasts such as osteocalcin (OCN), Dickkopf 1 (DKK1), and CCAAT/enhancer-binding protein beta (C/EBPß) were dramatically suppressed by TGFß1 treatment. The suppressive effects of TGFß1 were reversed with anti-TGFß1 treatment. Mechanically, TGFß1 decreased protein levels of C/EBPß without changing mRNA levels and reduced both mRNA and protein levels of DKK1. The degradation of the C/EBPß protein by TGFß1 was dependent on the ubiquitin-proteasome pathway. TGFß1 degraded the C/EBPß protein by inducing the expression of the E3 ubiquitin ligase Smad ubiquitin regulatory factor 1 (SMURF1) at the transcript level, thereby reducing the C/EBPß-DKK1 regulatory mechanism. Collectively, our findings suggest that TGFß1 suppressed the matrix mineralization of osteoblast differentiation by regulating the SMURF1-C/EBPß-DKK1 axis.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Calcificação Fisiológica/efeitos dos fármacos , Diferenciação Celular , Matriz Extracelular/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Osteoblastos/citologia , Fator de Crescimento Transformador beta1/farmacologia , Ubiquitina-Proteína Ligases/metabolismo , Idoso , Proteína beta Intensificadora de Ligação a CCAAT/genética , Matriz Extracelular/metabolismo , Regulação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteogênese , Ubiquitina-Proteína Ligases/genética
2.
Int Immunopharmacol ; 118: 110132, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37023698

RESUMO

OBJECTIVE: Ankylosing spondylitis (AS) exhibits paradoxical bone features typically characterized by new bone formation and systemic bone loss. Although abnormal kynurenine (Kyn), a tryptophan metabolite, has been closely linked to the disease activity of AS, the distinct role of its pathological bone features remains unknown. METHODS: Kynurenine sera level was collected from healthy control (HC; n = 22) and AS (n = 87) patients and measured by ELISA. In the AS group, we analyzed and compared the Kyn level based on the modified stoke ankylosing spondylitis spinal score (mSASSS), MMP13, and OCN. Under osteoblast differentiation, the treatment with Kyn in AS-osteoprogenitors conducted cell proliferation, alkaline phosphatase activity, bone mineralization-related alizarin red s (ARS), von kossa (VON), hydroxyapatite (HA) staining, and mRNA expression markers (ALP, RUNX2, OCN, and OPG) for bone formation. TRAP and F-actin staining was used for osteoclast formation of mouse osteoclast precursors. RESULTS: Kyn sera level was significantly elevated in the AS group compared to the HC. In addition, Kyn sera level was correlated with mSASSS (r = 0.03888, p = 0.067), MMP13 (r = 0.0327, p = 0.093), and OCN (r = 0.0436, p = 0.052). During osteoblast differentiation, treatment with Kyn exhibited no difference in cell proliferation and alkaline phosphate (ALP) activity for bone matrix maturation but promoted ARS, VON, and HA staining for bone mineralization. Interestingly, osteoprotegerin (OPG) and OCN expressions of AS-osteoprogenitors were augmented in the Kyn treatment during differentiation. In growth medium, Kyn treatment of AS-osteoprogenitors resulted in induction of OPG mRNA, protein expression, and Kyn-response genes (AhRR, CYP1b1, and TIPARP). Secreted OPG proteins were observed in the supernatant of AS-osteoprogenitors treated with Kyn. Notably, the supernatant of Kyn-treated AS-osteoprogenitors interrupted the RANKL-mediated osteoclastogenesis of mouse osteoclast precursor such as TRAP-positive osteoclast formation, NFATc1 expression, and osteoclast differentiation markers. CONCLUSION: Our results revealed that elevated Kyn level increased the bone mineralization of osteoblast differentiation in AS and decreased RANKL-mediated osteoclast differentiation by inducing OPG expression. Out study have implication for potential coupling factors linking osteoclast and osteoblast where abnormal Kyn level could be involved in pathological bone features of AS.


Assuntos
Cinurenina , Espondilite Anquilosante , Animais , Camundongos , Cinurenina/metabolismo , Metaloproteinase 13 da Matriz/metabolismo , Osteoblastos/metabolismo , Regulação da Expressão Gênica , Espondilite Anquilosante/metabolismo , Osteoclastos/metabolismo , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Diferenciação Celular , RNA Mensageiro/metabolismo , Ligante RANK/metabolismo
3.
Cells ; 10(12)2021 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-34944003

RESUMO

Aryl-hydrocarbon receptor (AhR) is a ligand-activated transcription factor and regulates differentiation and function of various immune cells such as dendritic cells, Th17, and regulatory T cells. In recent studies, it was reported that AhR is involved in bone remodeling through regulating both osteoblasts and osteoclasts. However, the roles and mechanisms of AhR activation in human osteoclasts remain unknown. Here we show that AhR is involved in human osteoclast differentiation. We found that AhR expressed highly in the early stage of osteoclastogenesis and decreased in mature osteoclasts. Kynurenine (Kyn), formylindolo[3,4-b] carbazole (FICZ), and benzopyrene (BaP), which are AhR agonists, inhibited osteoclast formation and Kyn suppressed osteoclast differentiation at an early stage. Furthermore, blockade of AhR signaling through CH223191, an AhR antagonist, and knockdown of AhR expression reversed Kyn-induced inhibition of osteoclast differentiation. Overall, our study is the first report that AhR negatively regulates human osteoclast differentiation and suggests that AhR could be good therapeutic molecule to prevent bone destruction in chronic inflammatory diseases such as rheumatoid arthritis (RA).


Assuntos
Diferenciação Celular , Cinurenina/farmacologia , Osteoclastos/citologia , Osteoclastos/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Transdução de Sinais , Artrite Reumatoide/patologia , Diferenciação Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Humanos , Receptores de Lipopolissacarídeos/metabolismo , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Receptores de Hidrocarboneto Arílico/agonistas , Líquido Sinovial/citologia
4.
Cell Death Dis ; 12(11): 971, 2021 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-34671026

RESUMO

While their function, as immune checkpoint molecules, is well known, B7-family proteins also function as regulatory molecules in bone remodeling. B7-H3 is a receptor ligand of the B7 family that functions primarily as a negative immune checkpoint. While the regulatory function of B7-H3 in osteoblast differentiation has been established, its role in osteoclast differentiation remains unclear. Here we show that B7-H3 is highly expressed in mature osteoclasts and that B7-H3 deficiency leads to the inhibition of osteoclastogenesis in human osteoclast precursors (OCPs). High-throughput transcriptomic analyses reveal that B7-H3 inhibition upregulates IFN signaling as well as IFN-inducible genes, including IDO. Pharmacological inhibition of type-I IFN and IDO knockdown leads to reversal of B7-H3-deficiency-mediated osteoclastogenesis suppression. Although synovial-fluid macrophages from rheumatoid-arthritis patients express B7-H3, inhibition of B7-H3 does not affect their osteoclastogenesis. Thus, our findings highlight B7-H3 as a physiologic positive regulator of osteoclast differentiation and implicate type-I IFN-IDO signaling as its downstream mechanism.


Assuntos
Antígenos B7/metabolismo , Diferenciação Celular , Indolamina-Pirrol 2,3,-Dioxigenase/biossíntese , Interferon Tipo I/metabolismo , Osteoclastos/metabolismo , Osteoclastos/patologia , Animais , Anticorpos Neutralizantes/farmacologia , Artrite Reumatoide/patologia , Antígenos B7/deficiência , Antígenos B7/genética , Indução Enzimática/efeitos dos fármacos , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Interferon beta/metabolismo , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Osteogênese/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Proteína 1 Supressora da Sinalização de Citocina/metabolismo , Líquido Sinovial/metabolismo , Triptofano/metabolismo
5.
Bone Res ; 9(1): 45, 2021 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-34671034

RESUMO

Osteoclasts are bone-resorbing cells that play an essential role in homeostatic bone remodeling and pathological bone erosion. Macrophage colony stimulating factor (M-CSF) is abundant in rheumatoid arthritis (RA). However, the role of M-CSF in arthritic bone erosion is not completely understood. Here, we show that M-CSF can promote osteoclastogenesis by triggering the proteolysis of c-FMS, a receptor for M-CSF, leading to the generation of FMS intracellular domain (FICD) fragments. Increased levels of FICD fragments positively regulated osteoclastogenesis but had no effect on inflammatory responses. Moreover, myeloid cell-specific FICD expression in mice resulted in significantly increased osteoclast-mediated bone resorption in an inflammatory arthritis model. The FICD formed a complex with DAP5, and the FICD/DAP5 axis promoted osteoclast differentiation by activating the MNK1/2/EIF4E pathway and enhancing NFATc1 protein expression. Moreover, targeting the MNK1/2 pathway diminished arthritic bone erosion. These results identified a novel role of c-FMS proteolysis in osteoclastogenesis and the pathogenesis of arthritic bone erosion.

6.
J Control Release ; 330: 1300-1312, 2021 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-33242532

RESUMO

Excessive tumor necrosis factor-α (TNF-α) is associated with the pathogenesis of rheumatoid arthritis (RA). Approximately 90% of patients with RA, who have inadequate response to methotrexate, follow anti-TNF-α therapy as the first-line immuno-treatment. However, ineffective long-term anti-TNF-α antibody cycling for 40% of non-responders to anti-TNF-α antibodies is costly and associated with various side effects, which needs alternative mechanism of action therapies. In the present study, a novel strategy to down-regulate TNF-α level was developed by using an alternative method of suppressing TNF-α converting enzyme (TACE), a transmembrane enzyme involved in cleaving and releasing bioactive soluble TNF-α. TACE suppression can be an effective remedy to block the production of soluble TNF-α, leading to an increased sensitivity to anti-TNF-α non-responders. A disease site-targeted RNA interference system was developed by forming non-viral complex between shRNA against TACE (shTACE) and bone resorption site-specific peptide carrier composed of aspartate repeating and arginine repeating sequences. The shTACE/peptide carrier complex alleviated arthritic symptoms in collagen induced arthritis (CIA) models by demonstrating enhanced anti-inflammatory and anti-osteoclastogenic effects. Similar results were obtained with human primary synovial cells and osteoclast precursor isolated from tissues and synovial fluids of RA patients. Taken together, the shTACE/targeting peptide complex provides a strong potential as an alternative anti-TNF-α therapeutic for RA treatment.


Assuntos
Artrite Experimental , Artrite Reumatoide , Proteína ADAM17 , Animais , Artrite Reumatoide/tratamento farmacológico , Humanos , Interferência de RNA , Inibidores do Fator de Necrose Tumoral , Fator de Necrose Tumoral alfa/metabolismo
7.
Immunol Lett ; 206: 33-40, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30543834

RESUMO

TGF-ß1 is highly expressed in the synovial tissue of patients with rheumatoid arthritis and is known as a cytokine that plays an important role in tissue repair and immune cell regulation. However, the role of TGF-ß1 is still unclear in osteoclastogenesis. In this study, we examined the effect of TGF-ß1 on osteoclast differentiation and the underlying mechanism using healthy human peripheral blood monocytes. TGF-ß1 was found to inhibit osteoclast differentiation and decrease the expression of osteoclast-specific genes such as acid phosphatase 5, tartrate resistant and cathepsin K. Levels of NFAT1, an important transcription factor in osteoclastogenesis, were also reduced. In addition, TGF-ß1 suppressed receptor activator of NF-κB (RANK) ligand-induced NF-κB and p38 MAPK signaling. Inhibition of osteoclast differentiation by TGF-ß1 was reversed by 1 µM SB431542 (an inhibitor of ALK4/5/7), which inhibited TGF-ß1-induced phosphorylation of SMAD1, but not that of SMAD3. TGF-ß1 also restricted RANK expression, and this was partially reversed by 1 µM SB431542. In contrast, the inhibition of SMAD3 by SIS3 (an inhibitor of SMAD3) reduced the osteoclast formation. TGF-ß1 has both inhibitory and stimulatory effects on human osteoclast differentiation, and that these opposing functions are mediated by SMAD1 and SMAD3 signaling, respectively.


Assuntos
Diferenciação Celular , Osteoclastos/citologia , Osteoclastos/metabolismo , Transdução de Sinais , Proteína Smad1/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Biomarcadores , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Expressão Gênica , Humanos , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Osteoclastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Ligante RANK/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Smad1/genética , Proteína Smad3/genética , Fator de Crescimento Transformador beta/farmacologia
8.
BMB Rep ; 52(6): 391-396, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30355436

RESUMO

Receptor activator of nuclear factor kappa B ligand (RANKL) expression in osteoblasts is regulated by 1,25-dihydroxyvitamin D3 (1,25D3). CCAAT/enhancer-binding protein beta (C/EBPß) has been proposed to function as a transcription factor and upregulate RANKL expression, but it is still uncertain how C/EBPß is involved in 1,25D3-induced RANKL expression of osteoblasts. 1,25D3 stimulation increased the expression of RANKL and C/EPBß genes in osteoblasts and enhanced phosphorylation and stability of these proteins. Moreover, induction of RANKL expression by 1,25D3 in osteoblasts was downregulated upon knockdown of C/EBPß. In contrast, C/EBPß overexpression directly upregulated RANKL promoter activity and exhibited a synergistic effect on 1,25D3-induced RANKL expression. In particular, 1,25D3 treatment of osteoblasts increased C/EBPß protein binding to the RANKL promoter. In conclusion, C/EBPß is required for induction of RANKL by 1,25D3. [BMB Reports 2019; 52(6): 391-396].


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Calcitriol/farmacologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Ligante RANK/metabolismo , Sítios de Ligação , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Núcleo Celular/metabolismo , Humanos , NF-kappa B/metabolismo , Osteoblastos/citologia , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas , Ligação Proteica , Estabilidade Proteica , Ligante RANK/biossíntese , Ligante RANK/genética , RNA Mensageiro/metabolismo
9.
Cell Death Differ ; 25(2): 432-443, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29099484

RESUMO

Staufen1 (STAU1) and Lin28B are RNA-binding proteins that are involved in neuronal differentiation as a function of post-transcriptional regulation. STAU1 triggers post-transcriptional regulation, including mRNA export, mRNA relocation, translation and mRNA decay. Lin28B also has multiple functions in miRNA biogenesis and the regulation of translation. Here, we examined the connection between STAU1 and Lin28B and found that Lin28B regulates the abundance of STAU1 mRNA via miRNA maturation. Decreases in the expression of both STAU1 and Lin28B were observed during neuronal differentiation. Depletion of STAU1 or Lin28B inhibited neuronal differentiation, and overexpression of STAU1 or Lin28B enhanced neuronal differentiation. Interestingly, the stability of STAU1 mRNA was modulated by miR-142-3p, whose maturation was regulated by Lin28B. Thus, miR-142-3p expression increased as Lin28B expression decreased during differentiation, leading to the reduction of STAU1 expression. The transcriptome from Staufen-mediated mRNA decay (SMD) targets during differentiation was analyzed, confirming that STAU1 was a key factor in neuronal differentiation. In support of this finding, regulation of STAU1 expression in mouse neural precursor cells had the same effects on neuronal differentiation as it did in human neuroblastoma cells. These results revealed the collaboration of two RNA-binding proteins, STAU1 and Lin28B, as a regulatory mechanism in neuronal differentiation.


Assuntos
Diferenciação Celular , Proteínas do Citoesqueleto/genética , MicroRNAs/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Animais , Proteínas do Citoesqueleto/metabolismo , Células HeLa , Humanos , Camundongos , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa