serum hmgb1 level
Recently Published Documents


TOTAL DOCUMENTS

10
(FIVE YEARS 4)

H-INDEX

3
(FIVE YEARS 1)

2021 ◽  
Vol 129 (Suppl_1) ◽  
Author(s):  
Jiandong Zhang ◽  
Peyton Sandroni ◽  
Wei Huang ◽  
Brian C Jensen

Our previous work has demonstrated essential protective roles for the endogenous cardiomyocyte alpha-1A adrenergic receptor (α1A-AR) subtype in mouse models of heart failure. However, the underlying mechanism of this protective phenotype is unclear. To address this gap in knowledge, we bred a mouse line lacking α1A-ARs on cardiomyocytes by crossing αMHC-cre mice with floxed α1A mice (CMKO= cre+ fl/fl, CMWT= cre- fl/fl), and subjected males to permanent LAD ligation. CMKO mice had increased serum HMGB1 level, larger infarcts and higher mortality. We found that RIP1/3-mediated programmed necrosis (necroptosis), but not apoptosis was exaggerated in CMKO mice 3 days after ligation. We then tested whether RIP1 inhibition with Nec-1s could mitigate this injury. Mice were given Nec-1s (1.65 mg/kg) or vehicle 10 mins prior to LAD ligation, followed by daily IV injection. Nec-1s treatment diminished post-ligation RIP1 (0.62±0.02 vs. 0.78±0.23 A.U., p=NS) and RIP3 expression (0.33±0.1 vs. 0.26±0.10 A.U., p=NS) in CMWT and CMKO mice respectively. Serum level of HMGB1 on D3 was markedly reduced in both CMWT (45.1%) and CMKO (61.1 %) after Nec-1s treatment. There was no difference between Nec-1s treated CMWT and CMKO mice (147±53 vs. 174±37 pg/mL, p=NS), indicating that blocking the RIP kinase pathway abrogates the exaggerated cell death in CMKO mice after ligation. Likewise, Nec-1s-treated CMKO mice had similar infarct areas to CMWT controls (16.2±4.5 vs. 19.9±4.6%, p=NS), further confirming that targeting necroptosis abrogates pathological damage. Collectively these Nec-1s data suggest that RIP-mediated necroptosis may account for larger infarcts in CMKO mice. Interestingly, expression of the apoptosis markers c-caspase-3 and PARP was similar between CMWT and CMKO mice, suggesting that the α1A-AR specifically regulates necroptosis. In sum, our data demonstrate that RIP kinase-mediated necroptosis contributes to susceptibility to injury in mice lacking cardiomyocyte α1A-ARs.


Author(s):  
Khalid S. Salih ◽  
Farqad B. Hamdan ◽  
Qasim S. Al-Mayah

Abstract Introduction There are large numbers of inflammatory molecules and humoral mediators that can be involved in the epileptogenesis such as cytokines, matrix metalloproteinases (MMP), and high mobility group box-1 (HMGB1). We aimed to evaluate serum levels and the diagnostic value of MMP-2 and HMGB1 in Iraqi patients with epilepsy. Methods One hundred epileptic patients comprised 60 controlled epileptics and 40 refractory patients to treatment with multi antiepileptic drugs (AEDs). Other 50 family-unrelated age- and sex-matched healthy subjects were selected to represent the control group. Serum levels of MMP-2 and HMGB1 were estimated using ELISA. The receiver operating characteristic (ROC) curve was used to evaluate the diagnostic value of these markers when required. Results MMP-2 level was significantly higher in controls than epileptic patients in general (controlled and refractory patients). ROC curve, showed poor diagnostic value of MMP-2 in discriminating epileptics into responsive or refractory to treatment from controls (AUC = 0.679 (95% CI = 0.536-0.823), and AUC = 0.77 (95% CI = 0.637-902), respectively). Serum HMGB1 level in epileptic patients and controls was in close approximation to each other. Conclusions MMP-2 is significantly decreased in patients particularly those with refractory epilepsy (RE); however, it has poor diagnostic value. No difference in the serum HMGB1 level between epileptic patients and controls.


2020 ◽  
Vol 21 (14) ◽  
pp. 5128
Author(s):  
Olga Troitskaya ◽  
Ekaterina Golubitskaya ◽  
Mikhail Biryukov ◽  
Mikhail Varlamov ◽  
Pavel Gugin ◽  
...  

The application of cold atmospheric plasma (CAP) in cancer therapy could be one of the new anticancer strategies. In the current work, we used cold atmospheric plasma jet for the treatment of cultured cells and mice. We showed that CAP induced the death of MX−7 mouse rhabdomyosarcoma cells with the hallmarks of immunogenic cell death (ICD): calreticulin and heat shock protein 70 (HSP70) externalization and high-mobility group box 1 protein (HMGB1) release. The intensity of HMGB1 release after the CAP treatment correlated directly with the basal extracellular HMGB1 level. Releasing from dying cells, HMGB1 can act as a proinflammatory cytokine. Our in vivo study demonstrated that cold atmospheric plasma induces a short-term two-times increase in serum HMGB1 level only in tumor-bearing mice with no effect in healthy mice. These findings support our hypothesis that CAP-dependent HMGB1 release from dying cancer cells can change the serum HMGB1 level. At the same time, we showed a weak cytokine response to CAP irradiation in healthy mice that can characterize CAP as an immune-safety physical antitumor approach.


2019 ◽  
Vol 9 (1) ◽  
Author(s):  
Feng Chen ◽  
Zuolong Liu ◽  
Wei Li ◽  
Dan Li ◽  
Bailing Yan

2017 ◽  
Vol 2017 ◽  
pp. 1-11 ◽  
Author(s):  
Zhilin Qi ◽  
Yao Zhang ◽  
Shimei Qi ◽  
Liefeng Ling ◽  
Lin Gui ◽  
...  

HMGB1, a highly conserved nonhistone DNA-binding protein, plays an important role in inflammatory diseases. Once released to the extracellular space, HMGB1 acts as a proinflammatory cytokine that triggers inflammatory reaction. Our previous study showed that salidroside exerts anti-inflammatory effect via inhibiting the JAK2-STAT3 signalling pathway. However, whether salidroside inhibits the release of HMGB1 is still unclear. In this study, we aim to study the effects of salidroside on HMGB1 release and then investigate the potential molecular mechanisms. In an experimental rat model of sepsis caused by CLP, salidroside administration significantly attenuated lung injury and reduced the serum HMGB1 level. In RAW264.7 cells, we investigated the effects of salidroside on LPS-induced HMGB1 release and then explored the underlying molecular mechanisms. We found that salidroside significantly inhibited LPS-induced HMGB1 release, and the inhibitory effect was correlated with the HMGB1 acetylation levels. Mechanismly, salidroside inhibits HMGB1 acetylation through the AMPK-SirT1 pathway. In addition, SirT1 overexpression attenuated LPS-induced HMGB1 acetylation and nucleocytoplasmic translocation. Furthermore, in SirT1 shRNA plasmid-transfected cells, salidroside treatment enhanced SirT1 expression and reduced LPS-activated HMGB1 acetylation and nucleocytoplasmic translocation. Collectively, these results demonstrated that salidroside might reduce HMGB1 release through the AMPK-SirT1 signalling pathway and suppress HMGB1 acetylation and nucleocytoplasmic translocation.


2015 ◽  
Vol 184 ◽  
pp. 318-320 ◽  
Author(s):  
Tao Liu ◽  
De-Yong Zhang ◽  
Yan-Hong Zhou ◽  
Qian-Feng Han ◽  
Lan-Hua Wang ◽  
...  

Circulation ◽  
2007 ◽  
Vol 116 (suppl_16) ◽  
Author(s):  
Takashi Kohno ◽  
Toshihisa Anzai ◽  
Tsutomu Yoshikawa ◽  
Kotaro Naito ◽  
Koji Ueno ◽  
...  

Background: The appropriate infarct healing process is prerequisite for preventing left ventricular (LV) remodeling after myocardial infarction (MI). High mobility group box1 protein (HMGB1) is derived from necrotic cells and activated macrophages and involved in tissue repair as immunostimulatory signal. However, the role of HMGB1 in post-MI LV remodeling is unknown. Methods and Results: We examined 35 patients with first ST-elevated acute MI. Peak serum HMGB1 level, determined by serial measurements (q. 6h), did not differ in age, sex, coronary risk factors and infarct site. Its peak level was markedly higher in patients with adverse in-hospital clinical outcomes, including pump failure, cardiac rupture, LV aneurysm and cardiac death, compared with those without (all p<0.05). Then we performed experimental study using rat MI model, induced by permanent left coronary ligation. Anti-HMGB1 antibodies (1 mg/day, MI/antiH, n=20) or control antibodies (MI/C, n=20) were injected subcutaneously every 24 hours for 7 days. Sham-operated rats served as controls (n=10). Quantitative RT-PCR and immunoblotting showed that HMGB1 expression was increased in the infarcted area peaking at day 3 in MI/C compared with sham (p<0.05). Its expression was localized in myocytes and inflammatory cells by immunohistochemistry. The mRNA level of TNF-α (−48%, p=0.03) and IL1-β (−44%, p=0.02) and the number of infiltrated macrophages (p=0.04) in the infarcted area were markedly reduced at day 3 in MI/antiH compared with MI/C. Echocardiographic and hemodynamic studies at day 7 showed increased LV end-diastolic dimension (p=0.03), LVEDP (p=0.03), and decreased LV max dP/dt (p=0.01) in MI/antiH compared with MI/C. Histologically, scar thinning in the infarcted area (0.53±0.23 vs 0.68±0.21 mm, p=0.02) and hypertrophy in the non-infarcted myocardium (2.01±0.23 vs 1.63±0.21 mm, p=0.03) were more prominent in MI/antiH compared with MI/C at day 14. Conclusions: Elevation of serum HMGB1 level is associated with adverse clinical outcomes in patients with acute MI. However, blockade of HMGB1 in rat MI model aggravated early LV remodeling through impaired infarct healing and advanced scar thinning, suggesting an essential role of HMGB1 in the appropriate repairing process after MI.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3827-3827
Author(s):  
Takashi Ito ◽  
Kazuhiro Abeyama ◽  
Ko-ichi Kawahara ◽  
Kamal K. Biswas ◽  
Tomonori Uchimura ◽  
...  

Abstract High Mobility Group box 1(HMGB1) is an abundant DNA-binding protein that acts as a proinflammatory cytokine when released in the extracellular milieu by necrotic and inflammatory cells. Moreover, an increased HMGB1 in the circulation of septic patients may induce multi-organ failure and lethality. However, very recent observations suggest that the protein also acts as an innate adjuvant, stem cell chemoattractant and growth factor. Thus only systemic and circulatory HMGB1 may induce morbidity and mortality, however, localized HMGB1 may have beneficial effects. Therefore, we serially examined the serum HMGB1 level in patients with various diseases, and also evaluated the significance of the protein. We demonstrate here how HMGB1 is localized and acts as an immune-adjuvant and a repairing factor in damaged tissue. We first established specific ELISA method to measure HMGB1. An increased level of HMGB1 was detected in the serum from patients with sever sepsis, infections, malignancy and so on. However, serum HMGB1 concentrations were fluctuated during the clinical course, and could not be concluded as a lethal mediator as previously reported. Next we investigated the reason of dynamic fluctuations of the protein in the circulation. Based on our findings, we proposed that this fluctuation of HMGB1 concentrations may be mediated by at least following three fashions; 1) proteolytic degradation by plasmin and thrombin, 2) endothelial thrombomodulin(TM) adsorption, and 3) generation of antibody against the protein. We observed that plasmin efficiently degraded HMGB1 into small fragments. However, interestingly the generated fragments of the protein still possess an ability to produce TNFa in macrophages through an undefined pathway. TM binds the protein on its N-terminus lectin-like domain. Binding of HMGB1 to TM resulted in decrement of TM’s cofactor activity to activate protein C by thrombin. HMGB1 bound to TM was gradually degraded by thrombin. These may be a system to localize HMGB1 only in injury sites where TM is down-regulated or disappeared through endothelial-loss. This may exert endothelial defense system against extracellular HMGB1 in severe tissue injury. Another possibility is that the generated antibody against HMGB1 may neutralize the proinflammatory action of the protein. In this context, we found that some of the antibodies against HMGB1 have the characteristics of P-ANCA(perinuclear anti-neutrophil cytoplasmic antibody). This may alter the phenotype of the underlining diseases. In conclusion, we suggest that HMGB1 is not merely a lethal mediator, but a kind of “testament” mediator of cell necrosis or invasive attacks to dendritic cells.


Sign in / Sign up

Export Citation Format

Share Document