hmgb1 expression
Recently Published Documents


TOTAL DOCUMENTS

164
(FIVE YEARS 61)

H-INDEX

21
(FIVE YEARS 4)

2021 ◽  
Author(s):  
Changhe Hou ◽  
Ming Lu ◽  
Zixiong Lei ◽  
Shuangwu Dai ◽  
Wei Chen ◽  
...  

Abstract Background Numerous studies have demonstrated the important roles of tumor-associated macrophages (TAMs) in osteosarcoma metastasis. In osteosarcoma, higher levels of HMGB1 correlate with osteosarcoma progression. However, whether HMGB1 is involved in the polarization of M2 macrophages into M1 macrophages in osteosarcoma still remains largely unknown. Methods HMGB1 and CD206 mRNA expression was measured by qRT-PCR in osteosarcoma tissues and cells. HMGB1 and RAGE protein expression was measured by western blotting. Osteosarcoma migration was measured using a Transwell and wound-healing assay. Osteosarcoma invasion was measured using a Transwell assay. Macrophage subtypes were detected using flow cytometry. Results HMGB1 is aberrantly overexpressed in osteosarcoma, and positively correlates with the TNM III & IV stages, lymph node metastasis, and distant metastasis. Silencing HMGB1 inhibits migration, invasion, and metastasis-related proteins in osteosarcoma cells. Furthermore, the reduced HMGB1 expression in the conditioned media derived from osteosarcoma cells also induces the polarization of M2 TAMs to M1 TAMs. In addition, silencing HMGB1 inhibits the liver and lung metastases of osteosarcoma and reduces the expression of HMGB1, CD163, and CD206 in vivo experiments. HMGB1 regulates macrophage polarization through RAGE. Interestingly, the polarized M2 macrophages could induce osteosarcoma migration and invasion, which in turn results in activation of HMGB1 expression in osteosarcoma cells to form a positive feedback loop. Conclusions HMGB1 and M2 macrophages enhance osteosarcoma migration, invasion, and metastasis capability through positive feedback regulation. These findings reveal the significance of tumor cell and TAM interaction in the metastatic microenvironment.


2021 ◽  
Author(s):  
Yan Yuan ◽  
Yuanxia Liu ◽  
Huijing Ye ◽  
Yuchen Feng ◽  
Zhenzhen Liu ◽  
...  

Abstract Diabetic kidney disease (DKD) is the major cause of end-stage renal disease and is closely associated with the inflammation. The aim of this study was to investigate the involvement of HMGB upon TLR2/IL-1β signaling pathway in diabetic renal injury. After intraperitoneal injection of STZ for wo consecutive days, mice developed DKD companying with remarkable renal injury, manifested with albuminuria and renal histological characteristics. In the kidney, the expressions of HMGB1, TLR2 and p-NF-κB were obviously increased, and the levels of TNF-α and IL-6 were significantly higher in DKD mice than that in normal mice. The infiltration of macrophages demonstrated by high stain of F4/80 and CD14 in the kidney of DKD mice. Furthermore, fibrosis related marker of Collagen IV, fibronectin and TGF-β1 showed highly expressions and accumulated in the kidney of DKD mice. In vitro, HMGB-mediated inflammatory pathway was activated in HMGB1/IL-1β-treated HK-2 cell, while C29 (a TLR2 inhibitor) could inhibited the HMGB1 expression. The result indicated that HMGB1-mediated TLR2/IL-1β signaling pathways facilitated macrophage recruitment and fibroblast proliferation, which resulting in a positive feedback to sustain a self-perpetuating cycle of renal inflammation. Therefore, HMGB1 might be a potential target in DKD therapeutic interventions.


2021 ◽  
pp. 108171
Author(s):  
Hui Chen ◽  
Gang Li ◽  
Jianqiang Zhang ◽  
Ting Zheng ◽  
Qianglin Chen ◽  
...  

2021 ◽  
Vol 11 ◽  
Author(s):  
Junhua Li ◽  
Wei Zhou ◽  
Qiang Mao ◽  
Dandan Gao ◽  
Lin Xiong ◽  
...  

Chemoresistance remains as a major hindrance in the treatment of hepatocellular carcinoma (HCC). High mobility group box protein 1 (HMGB1) enhances autophagic flux and protects tumor cells from apoptosis, which results in acquired drug resistance. However, the exact mechanisms underlying HMGB1-modulated autophagy in HCC chemoresistance remain to be defined. In the present study, we found that administration of doxorubicin (DOX) significantly promoted HMGB1 expression and induced HMGB1 cytoplasmic translocation in human HCC cell lines BEL7402 and SMMC7721, which enhanced autophagy that contributes to protecting HCC cells from apoptosis and increasing drug resistance. Moreover, we observed HMGB1 translocation and elevation of autophagy in DOX-resistant BEL7402 and SMMC7721 cells. Additionally, inhibition of HMGB1 and autophagy increased the sensitivities of BEL-7402 and SMMC-7721 cells to DOX and re-sensitized their DOX-resistant cells. Subsequently, we confirmed with HMGB1 regulated autophagy by activating the 5ʹ adenosine monophosphate-activated protein kinase (AMPK)/mTOR pathway. In summary, our results indicate that HMGB1 promotes acquired DOX resistance in DOX-treated BEL7402 and SMMC7721 cells by enhancing autophagy through the AMPK/mTOR signaling pathway. These findings provide the proof-of-concept that HMGB1 inhibitors might be an important targeted treatment strategy for HCC.


2021 ◽  
Vol 22 (20) ◽  
pp. 11276
Author(s):  
Fabian Essig ◽  
Lilith Babilon ◽  
Christoph Vollmuth ◽  
Alexander M. Kollikowski ◽  
Mirko Pham ◽  
...  

High-mobility group box 1 protein (HMGB1) is a damage-associated molecular pattern (DAMP) involved in neutrophil extracellular trap (NET) formation and thrombosis. NETs are regularly found in cerebral thromboemboli. We here analyzed associated HMGB1 expression in human thromboemboli retrieved via mechanical thrombectomy from 37 stroke patients with large vessel occlusion. HMGB1 was detected in all thromboemboli, accounting for 1.7% (IQR 0.6–6.2%) of the total thromboemboli area and was found to be colocalized with neutrophils and NETs and in spatial proximity to platelets. Correlation analysis revealed that the detection of HMGB1 was strongly related to the number of neutrophils (r = 0.58, p = 0.0002) and platelets (r = 0.51, p = 0.001). Our results demonstrate that HMGB1 is a substantial constituent of thromboemboli causing large vessel occlusion stroke.


2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Jianyuan Pan ◽  
Buse Alexan ◽  
Dorn Dennis ◽  
Chiristine Bettina ◽  
Laeuf Ilona Mariya Christoph ◽  
...  

Abstract Objective Little is known regarding the functional role of microRNA-193-3p (miR-193-3p) in sepsis. Hence, the aim of the present study was to investigate the effect of miR-193-3p on myocardial injury in mice with sepsis and its mechanism through the regulation of signal transducers and activators of transcription 3 (STAT3). Methods The mice model of sepsis was established by cecal ligation and puncture (CLP), septic mice were injected with miR-193-3p agomir, miR-193-3p antagomir or siRNA-STAT3. The expression of miR-193-3p, STAT3 and HMGB1 in the myocardial tissue of septic mice were detected. Cardiac ultrasound, hemodynamics, myocardial injury markers, inflammatory factors and cardiomyocyte apoptosis in septic mice were measured. Results MiR-193-3p expression was reduced while STAT3 expression was increased in septic mice. Down-regulated STAT3 or up-regulated miR-193-3p improved cardiac function, attenuated myocardial injury, inflammation and cardiomyocyte apoptosis in septic mice. Knockdown STAT3 reversed the role of inhibited miR-193-3p for mice with sepsis. miR-193-3p targeted STAT3, thereby inhibiting HMGB1 expression. Conclusion This study provides evidence that miR-193-3p targets STAT3 expression to reduce HMGB1 expression, thereby reducing septic myocardial damage. MiR-193-3p might be a potential candidate marker and therapeutic target for sepsis.


Sign in / Sign up

Export Citation Format

Share Document