Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(4): 167082, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38367899

RESUMO

Hemorrhagic shock (HS) leads to intestinal damage and subsequent multiple organ dysfunction syndrome. Intestinal barrier dysfunction is the main cause of multiple organ failure associated with HS. Leukocyte immunoglobulin-like receptor B4 (Lilrb4) belongs to the Ig superfamily and is a vital natural immunomodulatory receptor. The purpose of this study was to identify the role and molecular mechanism of Lilrb4 in HS-induced ileal injury. In this work, HS was established by femoral artery cannula and 90 min of HS (blood pressure, 35-40 mmHg), followed by resuscitation. RNA sequencing analysis showed that Lilrb4 was highly expressed in the ileum of HS rats. As observed, HS rats exhibited severe ileal injury, characterized by enlarged subepithelial space, edema, exfoliation and extensive loss of villi. Whereas, lentivirus system-mediated Lilrb4 overexpression considerably mitigated these alterations. HS led to increased release of markers associated with intestinal injury, which was effectively reversed by Lilrb4 overexpression. In addition, after resuscitation, Lilrb4 overexpression inhibited HS-triggered inflammatory response, as evidenced by decreased levels of proinflammatory cytokines. Lilrb4 also inhibited the activation of NF-κB signal induced by HS. Notably, Lilrb4 modulated the balance of regulatory T (Treg)-T helper 17 (Th17) cells in the mesenteric lymph node (MLN), which may also contribute to its protective role in HS progression. In aggregate, these findings confirmed that Lilrb4 overexpression protected against ileal injury caused by HS, indicating that Lilrb4 may be a potential candidate for the treatment of HS.


Assuntos
NF-kappa B , Choque Hemorrágico , Animais , Ratos , Íleo/metabolismo , NF-kappa B/metabolismo , Choque Hemorrágico/complicações , Transdução de Sinais
2.
FASEB J ; 38(1): e23334, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38050647

RESUMO

Mesenchymal stem cells (MSCs) are a popular cell source for repairing the liver. Improving the survival rate and colonization time of MSCs may significantly improve the therapeutic outcomes of MSCs. Studies showed that 78-kDa glucose-regulated protein (GRP78) expression improves cell viability and migration. This study aims to examine whether GRP78 overexpression improves the efficacy of rat bone marrow-derived MSCs (rBMSCs) in HS-induced liver damage. Bone marrow was isolated from the femurs and tibias of rats. rBMSCs were transfected with a GFP-labeled GRP78 expression vector. Flow cytometry, transwell invasion assay, scratch assay immunoblotting, TUNEL assay, MTT assay, and ELISA were carried out. The results showed that GRP78 overexpression enhanced the migration and invasion of rBMSCs. Moreover, GRP78-overexpressing rBMSCs relieved liver damage, repressed liver oxidative stress, and inhibited apoptosis. We found that overexpression of GRP78 in rBMSCs inhibited activation of the NLRP3 inflammasome, significantly decreased the levels of inflammatory factors, and decreased the expression of CD68. Notably, GRP78 overexpression activated the Nrf-2/HO-1 pathway and inhibited the NF-κB pathway. High expression of GRP78 efficiently enhanced the effect of rBMSC therapy. GRP78 may be a potential target to improve the therapeutic efficacy of BMSCs.


Assuntos
Doença Hepática Crônica Induzida por Substâncias e Drogas , Chaperona BiP do Retículo Endoplasmático , Células-Tronco Mesenquimais , Choque Hemorrágico , Animais , Ratos , Doença Hepática Crônica Induzida por Substâncias e Drogas/metabolismo , Células-Tronco Mesenquimais/metabolismo , NF-kappa B/metabolismo , Choque Hemorrágico/metabolismo
3.
Cell Signal ; 112: 110928, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37844714

RESUMO

Heat shock proteins (HSP) are a large family of peptide proteins that are widely found in cells. Studies have shown that the expression and function of HSPs in cells are very complex, and they can participate in cellular physiological and pathological processes through multiple pathways. Multiple heat shock proteins are associated with cancer cell growth, proliferation, metastasis, and resistance to anticancer drugs, and they play a key role in cancer development by ensuring the correct folding or degradation of proteins in cancer cells. As research hotspots, HSP90, HSP70 and HSP27 have been extensively studied in cancer so far. However, HSP20, also referred to as HSPB6, as a member of the small heat shock protein family, has been shown to play an important role in the cardiovascular system, but little research has been conducted on HSP20 in cancer. This review summarizes the current cellular functions of HSP20 in different cancer types, as well as its effects on cancer proliferation, progression, prognosis, and its other functions in cancer, to illustrate the close association between HSP20 and cancer. We show that, unlike most HSPs, HSP20 mainly plays an active anticancer role in cancer development, which is expected to provide new ideas and help for cancer diagnosis and treatment and research.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Proteínas de Choque Térmico/metabolismo , Neoplasias/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico HSP27/metabolismo , Proteínas de Choque Térmico HSP20/metabolismo
4.
Oncol Lett ; 24(6): 462, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36380873

RESUMO

Heat shock protein (HSP) 20 belongs to the small HSP family and exhibits diverse functions, including tumor suppression, in addition to being a molecular chaperon, which is the classical property of HSPs. The present study aimed to examine the association between HSP20 expression and breast cancer (BC) progression in patients, and to explore the possible role of HSP20 in malignant phenotypes of BC cells. A series of experiments, including reverse transcription-quantitative PCR, western blotting, Cell Counting Kit-8 and flow cytometry, were performed. Data from Gene Expression Omnibus and Kaplan-Meier Plotter revealed that HSP20 expression was significantly downregulated in BC tissues, and patients with BC with lower HSP20 expression exhibited poorer recurrence-free survival. The data revealed that HSP20 was closely associated with the pathological tumor stage (P=0.015) and pathological tumor node metastasis (P=0.031) of patients with BC. Additionally, HSP20 expression was markedly decreased in BC cell lines. Exogenous overexpression of HSP20 inhibited proliferation and accelerated apoptosis of BC cells. These cells exhibited decreased migration and invasion when HSP20 was overexpressed. Furthermore, HSP20 overexpression suppressed the MAPK and AKT signaling pathways, as evidenced by the reduced phosphorylation levels of AKT, ERK, JNK and p38. Knockdown of HSP20 exerted the opposite effects. Notably, the AKT agonist, SC79, and the ERK agonist, LM22B-10, reversed the decrease in cell proliferation and migration induced by HSP20 overexpression. Overall, the data suggest that the decreased expression of HSP20 in BC tissues may be associated with disease progression. HSP20 also attenuated the malignant phenotype of BC cells and the inhibition of MAPK and AKT signaling may be associated with this effect. Therefore, HSP20 may be a potential prognostic marker or a candidate therapeutic target for BC.

5.
J Cell Mol Med ; 25(24): 11075-11084, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34734476

RESUMO

Spinal cord injury (SCI), a major cause of disability, causes high global disease and economic burdens. Stress-induced phosphoprotein 1 (STIP1) has been identified to be involved in spinal cord ischaemia-reperfusion injury (SCII); however, the effect of STIP1 on SCII remains unclear until now. This study aimed to examine the role of STIP1 in SCII and unravel the possible mechanisms. Western blotting and immunohistochemical staining showed that STIP1 expression rapidly increased and then decreased in rat spinal cord following SCII treatment. Neurological function scoring, HE staining, immunohistochemical staining and Western blotting revealed that STIP1 overexpression alleviated SCII-induced motor dysfunction of hind limbs, neuronal loss and inflammation in spinal cord, and inhibited activity of nuclear factor kappa B (NF-κB) signalling in rats. Immunoprecipitation identified that STIP1 was co-located with Iba-1. In addition, STIP1 was found to ameliorate oxygen and glucose deprivation (OGD)-induced inflammation and activation of NF-κB signalling in mouse microglia BV2 cells, and STIP1 resulted in decrease of heat shock protein family A member 8 (HSPA8), increase of IκBß expression and reduced binding of IκBß to HSPA8 in BV2 cells. The results of the present study demonstrate that STIP1 alleviates ischaemia/reperfusion-induced neuronal injury and inflammation in rat spinal cord and mouse microglial cells by deactivating NF-κB signalling. These findings may provide novel insights for the clinical diagnosis and treatment of SCI.


Assuntos
Proteínas de Choque Térmico/genética , NF-kappa B/metabolismo , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais , Isquemia do Cordão Espinal/complicações , Animais , Modelos Animais de Doenças , Suscetibilidade a Doenças , Imunofluorescência , Expressão Gênica , Proteínas de Choque Térmico/metabolismo , Imuno-Histoquímica , Masculino , Microglia/metabolismo , Microglia/patologia , Atividade Motora , Neurônios/metabolismo , Ratos , Traumatismo por Reperfusão/patologia , Isquemia do Cordão Espinal/etiologia , Isquemia do Cordão Espinal/metabolismo , Isquemia do Cordão Espinal/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...