Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Bases de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Cent Eur J Immunol ; 48(1): 63-69, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37206587

RESUMO

Homeobox containing 1 (HMBOX1) is a transcription factor that was identified in 2006 from a cDNA library of the human pancreas. It belongs to the HNF gene class of the homeobox family. HMBOX1 is widely expressed in normal human tissues; however, its expression level is rather uneven. Homeobox members have been widely reported to participate in embryonic development and differentiation as well as in pathological and physiological processes. Although research on the role of HMBOX1 is still in its infancy, many reports have revealed its regulatory role in cell differentiation, immune regulation, inflammation, and tumor progression. HMBOX1 plays an important role in promoting the differentiation of bone marrow stromal stem cells (BMSCs) into endothelial cells and contributes to their physiological functions. As an immunoregulatory factor, HMBOX1 can significantly inhibit the inflammatory response in hepatocytes and NK cells and impede the infiltration of peripheral immune cells to the liver. In tumor development, HMBOX1 exerts diametrically opposite biological functions, inhibiting or promoting the process. HMBOX1 possesses complex and diverse biological functions. In this review, we provide a brief overview of the research on HMBOX1.

2.
J Cell Mol Med ; 23(1): 453-463, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30358079

RESUMO

Inflammation and reactive oxygen species (ROS) are important factors in the pathogenesis of atherosclerosis (AS). 5,2'-dibromo-2,4',5'-trihydroxydiphenylmethanone (TDD), possess anti-atherogenic properties; however, its underlying mechanism of action remains unclear. Therefore, we sought to understand the therapeutic molecular mechanism of TDD in inflammatory response and oxidative stress in EA.hy926 cells. Microarray analysis revealed that the expression of homeobox containing 1 (HMBOX1) was dramatically upregulated in TDD-treated EA.hy926 cells. According to the gene ontology (GO) analysis of microarray data, TDD significantly influenced the response to lipopolysaccharide (LPS); it suppressed the LPS-induced adhesion of monocytes to EA.hy926 cells. Simultaneously, TDD dose-dependently inhibited the production or expression of IL-6, IL-1ß, MCP-1, TNF-α, VCAM-1, ICAM-1 and E-selectin as well as ROS in LPS-stimulated EA.hy926 cells. HMBOX1 knockdown using RNA interference attenuated the anti-inflammatory and anti-oxidative effects of TDD. Furthermore, TDD inhibited LPS-induced NF-κB and MAPK activation in EA.hy926 cells, but this effect was abolished by HMBOX1 knockdown. Overall, these results demonstrate that TDD activates HMBOX1, which is an inducible protective mechanism that inhibits LPS-induced inflammation and ROS production in EA.hy926 cells by the subsequent inhibition of redox-sensitive NF-κB and MAPK activation. Our study suggested that TDD may be a potential novel agent for treating endothelial cells dysfunction in AS.


Assuntos
Anti-Inflamatórios/farmacologia , Proteínas de Homeodomínio/metabolismo , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Lipopolissacarídeos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Aterosclerose/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , Inflamação/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo
3.
Exp Ther Med ; 23(3): 224, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35222701

RESUMO

Homeobox containing 1 (HMBOX1) is a member of the homeobox transcription factor family that has been reported to serve an important role in numerous biological processes. The present study aimed to determine the role of HMBOX1 in the pathogenesis of periodontitis. Human periodontal ligament stem cells (hPDLSCs) were treated with liposaccharide (LPS) and transfected with a HMBOX1 overexpression (Ov-HMBOX1) plasmid or small interfering (si)-C-X-C motif chemokine ligand 10 (CXCL10) plasmids. The effects of Ov-HMBOX1 on cell proliferation, inflammation and apoptosis were subsequently investigated using Cell Counting Kit-8, ELISA for analysis of IL-6, TNF-α and IL-1ß levels, TUNEL and western blotting assays for analysis of Bcl-2, Bax, cleaved caspase-3 and caspase-3 levels, respectively. Furthermore, the potential effects of HMBOX1 on the mRNA and protein levels of CXCL10 and the NF-κB signaling pathway were investigated by using reverse transcription-quantitative PCR and western blotting. Finally, the physiological processes of lipopolysaccharide (LPS)-induced hPDLSCs overexpressing HMBOX1 were assessed following treatment with phorbol 12-myristate 13-acetate (PMA), a NF-κB agonist. The results revealed that Ov-HMBOX1 transfection promoted proliferation whilst alleviating inflammation and apoptosis in LPS-induced hPDLSCs. Ov-HMBOX1 reduced the expression of CXCL10 by suppressing the NF-κB signaling pathway. PMA treatment inhibited the proliferation of LPS-induced hPDLSCs transfected with Ov-HMBOX1, which was reversed by transfection with si-CXCL10. In conclusion, results of the present study provided evidence that HMBOX1 can attenuate LPS-induced hPDLSC injury by downregulating CXCL10 expression via the NF-κB signaling pathway, which may provide a novel insight into the development of potentially novel treatment strategies for periodontitis.

4.
Int J Biochem Cell Biol ; 79: 33-40, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27506770

RESUMO

Homeobox containing 1 (HMBOX1) is essential for the survival of human umbilical vein endothelial cells (HUVECs). However, the regulatory mechanism of HMBOX1 expression is still unclear. We recently found that a small molecule 6-amino-2,3-dihydro-3-hydroxymethyl-1,4-benzoxazine (ABO) directly targeted annexin A7 (ANXA7) and inhibited its GTPase activity. In addition, both HMBOX1 and ANXA7 participated in the autophagy and apoptosis of HUVECs. But, their relationship in the regulation of HMBOX1 expression is unknown. In this study, we found that ABO could elevate HMBOX1 at translation level through inhibiting ANXA7 GTPase activity. ABO failed to increase HMBOX1 protein level in ANXA7-deficient HUVECs. TGFB2 overlapping transcript 1 (TGFB2-OT1) that was increased by ABO facilitated HMBOX1 expression by increasing La-related protein 1 (LARP1) expression. Furthermore, the protein level of HMBOX1 was decreased under oxidized low-density lipoprotein (oxLDL) treatment in HUVECs and in the aortic endothelium of apolipoprotein E-deficient (apoE-/-) mice, which could be reversed by ABO in vitro and in vivo. In conclusion, ANXA7 was an endogenous regulator of HMBOX1, and ABO promoted HMBOX1 translation by inhibiting ANXA7 GTPase activity and enhancing TGFB2-OT1 expression. Besides, our data suggested that HMBOX1 might be a novel diagnostic marker and therapeutic target of atherosclerosis.


Assuntos
Anexina A7/antagonistas & inibidores , Benzoxazinas/farmacologia , GTP Fosfo-Hidrolases/metabolismo , Proteínas de Homeodomínio/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Homeodomínio/genética , Humanos , Masculino , Camundongos , Transporte Proteico/efeitos dos fármacos , RNA Longo não Codificante/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA