Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
1.
Avicenna J Med Biotechnol ; 16(2): 120-129, 2024.
Article in English | MEDLINE | ID: mdl-38618508

ABSTRACT

Background: Colorectal Cancer (CRC) represents a significant global health challenge, and its progression, resistance to therapy, and metastasis are strongly influenced by the tumor microenvironment, including factors like hypoxia. This study explores the impact of High Mobility Group Box 1 (HMGB1) overexpression on CRC cell migration, while identifying potential genes associated with this process. Methods: To explore this, we developed oncolytic virotherapy, resulting in HSVHMGB1, an oncolytic Herpes simplex virus that expresses HMGB1. HMGB1 is known its role in cancer progression, particularly in the context of cancer cell migration. Results: Contrary to expectations, our scratch assays indicated that HSV-HMGB1 did not significantly induce migration in CRC cells, suggesting that HMGB1 might not directly contribute to this process. Employing microarray analysis, we investigated gene expression changes linked to CRC cell migration, leading to construction of a Protein-Protein Interaction (PPI) network. This network revealed the presence of hub proteins, including as NDRG1, LGALS1, and ANGPTL4, which are recognized for their roles in cancer cell migration. The differential expression of these genes under hypoxic conditions was further validated using quantitative RT-PCR, aligning with the findings from our microarray data. Conclusion: Our findings emphasize the complex regulation of CRC cell migration, and provides valuable insights into potential molecular mechanisms and pathways. These findings have implications for further research into cancer progression and the development of therapeutic strategies.

2.
World J Mens Health ; 2024 Feb 19.
Article in English | MEDLINE | ID: mdl-38449454

ABSTRACT

PURPOSE: The primary goal of this study is to evaluate the effect of the non-invasive radiofrequency hyperthermia (RFHT) device on chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) rat model and investigate the underlying mechanism. MATERIALS AND METHODS: In this study, Sprague-Dawley rats were randomly distributed into three groups: (1) normal control group, (2) CP/CPPS group, and (3) RFHT group. CP/CPPS rat models were induced by 17ß-estradiol and dihydrotestosterone for 4 weeks and RFHT was administered for 5 weeks after model establishment. During RFHT administration, core body temperatures were continuously monitored with a rectal probe. After administering RFHT, we assessed pain index for all groups and collected prostate tissues for Western blot analysis, immunofluorescence, and immunohistochemistry. We also collected adjacent organs to the prostate including urinary bladder, testes, and rectum for safety assessment via H&E staining along with a terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling assay. RESULTS: After administering RFHT, pain in rats was significantly alleviated compared to the CP/CPPS group. RFHT reduced high-mobility group box 1 (HMGB1) expression and improved inflammation by downregulating subsequent proinflammatory cytokines through inhibition of the toll-like receptor 4 (TLR4)-nuclear factor kappa B (NF-κB) pathway. In prostate-adjacent organs, no significant histological alteration or inflammatory infiltration was detected. The area of cell death also did not increase significantly after RFHT. CONCLUSIONS: In conclusion, RFHT demonstrated anti-inflammatory effects by inhibiting the HMGB1-TLR4-NF-κB pathway in CP/CPPS rat models. This suggests that RFHT could serve as a safe and promising therapeutic strategy for CP/CPPS.

3.
Theranostics ; 13(12): 4059-4078, 2023.
Article in English | MEDLINE | ID: mdl-37554277

ABSTRACT

Rationale: CD93, a C-type lectin-like transmembrane glycoprotein, can be shed in a soluble form (sCD93) upon inflammatory stimuli. sCD93 effectively enhances apoptotic cell clearance and has been proposed as an inflammatory disease biomarker. The function of sCD93 involved directly in inflammation remains to be determined. Herein, we attempted to examine the hypothesis that sCD93 might sequester proinflammatory high-mobility group box 1 protein (HMGB1), exerting anti-inflammatory properties. Methods: Different forms of soluble recombinant human CD93 (rCD93) were prepared by a mammalian protein expression system. rCD93-HMGB1 interaction was assessed using co-immunoprecipitation and solid-phase binding assays. Effects of soluble rCD93 were evaluated in HMGB1-induced macrophage and vascular smooth muscle cells (VSMC) activation and receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclastogenesis, CaCl2-induced and angiotensin II-infused abdominal aortic aneurysm (AAA) formation and ovariectomized-induced osteoporosis in mice. Results: Protein binding studies revealed that soluble rCD93, via the lectin-like domain (D1), can bind to HMGB1 and intercept HMGB1-receptor interaction. Soluble rCD93 containing D1 inhibited HMGB1-induced proinflammatory cytokine production and intracellular mitogen-activated protein kinase (MAPK)/nuclear factor (NF)-κB activation in macrophages and VSMCs, thereby attenuating CaCl2-induced and angiotensin II-infused AAA models. During osteoclastogenesis, RANKL stimulated HMGB1 secretion that promoted RANKL-induced osteoclastogenesis in return. Soluble rCD93 containing D1 impeded RANKL-induced osteoclastogenic marker gene expression and intracellular MAPK/NF-κB signaling, thereby mitigating ovariectomized-induced osteoporosis. Conclusion: These findings demonstrate the therapeutic potential of soluble recombinant CD93 containing D1 in inflammatory diseases. Our study highlights a novel anti-inflammatory mechanism, i.e., sequestration of HMGB1, through which sCD93 prevents HMGB1-receptor interaction on effector cells and alleviates inflammation.


Subject(s)
HMGB1 Protein , Humans , Animals , Mice , HMGB1 Protein/metabolism , Lectins , Angiotensin II , Calcium Chloride , Inflammation , Mammals/metabolism
4.
Immune Netw ; 23(3): e26, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37416931

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection induces excessive pro-inflammatory cytokine release and cell death, leading to organ damage and mortality. High-mobility group box 1 (HMGB1) is one of the damage-associated molecular patterns that can be secreted by pro-inflammatory stimuli, including viral infections, and its excessive secretion levels are related to a variety of inflammatory diseases. Here, the aim of the study was to show that SARS-CoV-2 infection induced HMGB1 secretion via active and passive release. Active HMGB1 secretion was mediated by post-translational modifications, such as acetylation, phosphorylation, and oxidation in HEK293E/ACE2-C-GFP and Calu-3 cells during SARS-CoV-2 infection. Passive release of HMGB1 has been linked to various types of cell death; however, we demonstrated for the first time that PANoptosis, which integrates other cell death pathways, including pyroptosis, apoptosis, and necroptosis, is related to passive HMGB1 release during SARS-CoV-2 infection. In addition, cytoplasmic translocation and extracellular secretion or release of HMGB1 were confirmed via immunohistochemistry and immunofluorescence in the lung tissues of humans and angiotensin-converting enzyme 2-overexpressing mice infected with SARS-CoV-2.

5.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-992771

ABSTRACT

Objective:To investigate the role and underlying mechanisms of inhibiting high mobility group box-1 (HMGB1) in the expression of matrix metalloproteinase-9 (MMP-9) in spinal cord astrocytes (AS) in rats after spinal cord injury (SCI).Methods:After an SCI model was established in Sprague-Dawley (SD) rats using a modified Allen's Weight-Dropping method and ethyl pyruvate (EP) or glycyrrhizin (GL) was used to inhibit the effect of HMGB1, the rats were divided into a sham group, an SCI group, an SCI+EP (50 mg/kg) group, and an SCI+GL (100 mg/kg) group. The expression levels of glial fibrillary acid protein (GFAP) and MMP-9 in spinal cord AS were observed. After the spinal cord AS in SD rats was cultured and incubated by the oxygen-glucose deprivation/reoxygenation (OGD/R) procedure, the expression of MMP-9 protein was detected at 6 h/R 6 h, 12 h, 24 h, and 48 h after OGD. The time point with the highest expression was chosen in the subsequent experiments as an OGD/R group. HMGB1 was inhibited by HMGB1 shRNA or EP to observe the effect of HMGB1 on the expression of MMP-9 protein in AS treated with OGD/R. Then, toll-like receptor 4 (TLR4) inhibitor, TIR-domain-containing adaptor inducing interferon- β (TRIF) inhibitor, and nuclear factor-kappa B (NF- κB) inhibitor were used to investigate the effects of TLR4/TRIF/NF- κB signaling pathway during the regulation of HMGB1 on MMP-9 in vitro. Results:Western blot showed that the expression of MMP-9 protein in the spinal cord was significantly increased in rats at 1 d after SCI, and the expression of MMP-9 protein in the SCI+EP group and the SCI+GL group was significantly lower than that in the SCI group ( P<0.001). Immunofluorescence showed that GFAP and MMP-9 proteins were co-localized in the spinal cord after SCI, and the expression of GFAP and MMP-9 proteins in the SCI+EP and SCI+GL groups was significantly lower than that in the SCI group ( P<0.05). Since the expression of MMP-9 protein in the spinal cord AS cultured in vitro was significantly higher in the OGD 6h/R 12h group than that in the normal group and the OGD 6h/R 6h, 24, and 48 h groups, the OGD 6h/R 12h was taken as the OGD/R group. The MMP-9 protein expression in AS in the OGD/R+HMGB1 shRNA group and the OGD/R+EP group was significantly lower than that in the OGD/R group ( P<0.001). In the cultured AS, moreover, inhibiting TLR4, TRIF, and NF- κB reduced MMP-9 protein expression after OGD 6 h/R 12 h when compared with that in the OGD/R group ( P<0.001). Conclusions:HMGB1 inhibition may result in a reduction in MMP-9 expression both in the spinal cord AS in SCI rats and in AS after OGD/R treatment in vitro. HMGB1 may regulate MMP-9 protein expression in AS after OGD/R treatment via the TLR4/TRIF/NF- κB signal pathway.

6.
Acta Clin Croat ; 62(1): 25-35, 2023 Apr.
Article in English | MEDLINE | ID: mdl-38304371

ABSTRACT

Immunoglobulin A vasculitis (IgAV) or Henoch-Schönlein purpura is the most prevalent systemic small vessel vasculitis in childhood. High mobility group box 1 protein (HMBG1) is a pleiotropic cytokine that functions as a pro-inflammatory signal, important for the activation of antigen-presenting cells and propagation of inflammation. HMGB1 is implicated in the pathophysiology of a variety of inflammatory diseases. The aim of this study was to investigate the role of single nucleotide polymorphism rs41369348 for HMGB1 gene in the susceptibility and clinical features of patients meeting the classification criteria for IgAV. DNA was extracted from blood cells of 76 children with IgAV and 150 age-matched healthy controls. Clinical data and laboratory parameters were collected for all IgAV patients. Although there was a higher frequency of heterozygous A/delA genotype of this gene polymorphism in IgAV group as compared with control group, no genotype difference was observed between these two groups. No statistically significant genotype differences were disclosed when patients with different IgAV clinical features were compared. In conclusion, in this study, polymorphism rs41369348 for HMGB1 was not associated with increased susceptibility to childhood IgAV, its severity or different clinical manifestations.


Subject(s)
HMGB1 Protein , IgA Vasculitis , Vasculitis , Child , Humans , IgA Vasculitis/genetics , IgA Vasculitis/complications , HMGB1 Protein/genetics , Vasculitis/complications , Immunoglobulin A/genetics , Polymorphism, Single Nucleotide
7.
Braz. j. otorhinolaryngol. (Impr.) ; 88(6): 875-881, Nov.-Dec. 2022. tab, graf
Article in English | LILACS-Express | LILACS | ID: biblio-1420791

ABSTRACT

Abstract Introduction: Serum level of high-mobility group box 1 protein is reportedly correlated with the severity of obstructive sleep apnea. Objective: We tried to evaluate the possibility of using the serum high-mobility group box 1 protein level as a biologic marker in obstructive sleep apnea patients. Methods: We generated a chronic intermittent hypoxia murine model that reflected human obstructive sleep apnea. Obstructive sleep apnea patients who underwent polysomnography were prospectively enrolled. Serum samples were obtained from mice and obstructive sleep apnea patients, and the serum high-mobility group box1 protein level was measured by enzyme-linked immunosorbent assay. Results: Serum high-mobility group box 1 protein level was 56.16 ± 30.33 ng/mL in chronic intermittent hypoxia and 18.63 ± 6.20 ng/mL in control mice (p<0.05). The mean apnea-hypopnea index and respiratory disturbance index values of enrolled obstructive sleep apnea patients were 50.35 ± 27.96 and 51.56 ± 28.53, respectively, and the mean serum high-mobility group box 1 protein level was 30.13 ± 19.97 ng/mL. The apnea-hypopnea index and respiratory disturbance index were not significantly correlated with the serum high-mobility group box 1 protein level (p>0.05). Instead, this protein level was significantly correlated with lowest arterial oxygen concentration (SaO2) (p<0.05). Conclusion: High-mobility group box 1 protein may be involved in the pathogenesis of obstructive sleep apnea, and the possibility of this protein being a useful biologic marker in obstructive sleep apnea should be further evaluated.


Resumo Introdução: O nível sérico da proteína de alta mobilidade do grupo Box-1 está relacionado com a gravidade da apneia obstrutiva do sono. Objetivo: Avaliar o uso do nível sérico da proteína de alta mobilidade do grupo Box-1 como um marcador biológico em pacientes com apneia obstrutiva do sono. Método: Geramos um modelo murino de hipóxia intermitente crônica que imita a apneia obstrutiva do sono em humanos. Pacientes com apneia obstrutiva do sono que fizeram polissonografia foram incluídos prospectivamente. Amostras de soro foram obtidas de camundongos e pacientes com apneia obstrutiva do sono e o nível sérico da proteína de alta mobilidade do grupo Box-1 foi medido por enzyme-linked immunosorbent assay. Resultados: O nível sérico da proteína de alta mobilidade do grupo Box-1 foi 56,16 ± 30,33 ng/mL em hipóxia intermitente crônica e 18,63 ± 6,20 ng/mL em camundongos controle (p < 0,05). Os valores médios do índice de apneia-hipopneia e do índice de distúrbio respiratório nos pacientes com apneia obstrutiva do sono foram 50,35 ± 27,96 e 51,56 ± 28,53, respectivamente, e o nível médio da proteína de alta mobilidade do grupo Box-1 foi 30,13 ± 19,97 ng/mL. O índice de apneia-hipopneia e o índice de distúrbio respiratório não foram significantemente associados com o nível da proteína de alta mobilidade do grupo Box-1 p> 0,05). Em vez disso, esse nível de proteína foi significantemente associado com o valor mais baixo da concentração arterial de oxigênio (SaO2) (p <0,05). Conclusão: A proteína de alta mobilidade do grupo Box-1 pode estar envolvida na patogênese da apneia obstrutiva do sono e a possibilidade de que essa proteína possa ser um marcador biológico útil na apneia obstrutiva do sono deve ser avaliada mais detalhadamente.

8.
Anaesthesiologie ; 71(10): 741-749, 2022 10.
Article in German | MEDLINE | ID: mdl-36064976

ABSTRACT

BACKGROUND: Acutely occurring organ damage significantly contributes to morbidity and mortality in the perioperative context. OBJECTIVE: This article highlights new clinical perspectives on how perioperative organ damage can be prevented and ameliorated by influencing the high mobility group box 1 protein (HMGB1) signaling. MATERIAL AND METHODS: A MEDLINE search was performed in the fields of clinical and basic research. The presentation of basic mechanisms of perioperative organ damage and the discussion of the importance of HMGB1 in prevention and treatment by pharmaceutical and nonpharmaceutical interventions are the focus of the review. RESULTS: The HMGB1 is a central element in the pathogenesis of septic and aseptic inflammation-induced organ damage. Remote ischemic preconditioning (RIPC) and dexmedetomidine are highly effective approaches to mitigate or prevent organ damage. CONCLUSION: The RIPC and dexmedetomidine offer protective properties in ischemia-reperfusion injury as well as in inflammation-related organ damage, which are mediated by HMGB1, among others. This effectively protects the kidneys, heart, lungs, liver and brain. The application of these concepts should be considered in routine clinical practice.


Subject(s)
Dexmedetomidine , HMGB1 Protein , Ischemic Preconditioning , Dexmedetomidine/pharmacology , HMGB1 Protein/metabolism , Humans , Inflammation , Pharmaceutical Preparations
9.
Am J Physiol Cell Physiol ; 323(4): C1149-C1160, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36036451

ABSTRACT

High-mobility group box 1 (HMGB1) not only induces cell proliferation and migration but also promotes cell apoptosis and autophagy. Abnormal expression of HMGB1 in plasma or body fluids could be detected in the occurrence and development of inflammation, cardiovascular disease, immune diseases, and cancer. In recent years, the accumulating research on lncRNAs had led us to the important role of lncRNAs in finely regulating the expression of molecules. Nevertheless, the roles of lncRNAs in upregulating HMGB1 and its receptors remain elusive. This article systematically summarizes the lncRNAs related to HMGB1 and its essential receptors such as RAGE. Multiple lncRNAs, such as lncRNA MALAT1 were proposed to regulate HMGB1 and its receptors upstream. As HMGB1-related diseases were summarized, we also expected predictable application prospects of both HMGB1 and related lncRNAs. The in-depth research focusing on lncRNAs behind HMGB1 and its receptors might provide a novel foundation for therapeutic treatment of HMGB1-related disorders, together with targets regarding HMGB1.


Subject(s)
HMGB1 Protein , MicroRNAs , RNA, Long Noncoding , Cell Proliferation/genetics , HMGB1 Protein/genetics , MicroRNAs/genetics , Molecular Biology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
10.
Int J Mol Sci ; 23(6)2022 Mar 10.
Article in English | MEDLINE | ID: mdl-35328426

ABSTRACT

Owing to a rapid increase in aging population in recent years, the deterioration of motor function in older adults has become an important social problem, and several studies have aimed to investigate the mechanisms underlying muscle function decline. Furthermore, structural maintenance of the muscle-tendon-bone complexes in the muscle attachment sites is important for motor function, particularly for joints; however, the development and regeneration of these complexes have not been studied thoroughly and require further elucidation. Recent studies have provided insights into the roles of mesenchymal progenitors in the development and regeneration of muscles and myotendinous junctions. In particular, studies on muscles and myotendinous junctions have-through the use of the recently developed scRNA-seq-reported the presence of syncytia, thereby suggesting that fibroblasts may be transformed into myoblasts in a BMP-dependent manner. In addition, the high mobility group box 1-a DNA-binding protein found in nuclei-is reportedly involved in muscle regeneration. Furthermore, studies have identified several factors required for the formation of locomotor apparatuses, e.g., tenomodulin (Tnmd) and mohawk (Mkx), which are essential for tendon maturation.


Subject(s)
Muscle, Skeletal , Tendons , Cell-Matrix Junctions , Muscle Development/physiology , Muscle, Skeletal/metabolism , Myoblasts , Tendons/metabolism
11.
Acute Med Surg ; 9(1): e726, 2022.
Article in English | MEDLINE | ID: mdl-35127103

ABSTRACT

AIM: High mobility group box-1 (HMGB1) is a lethal mediator of sepsis that binds to haptoglobin (Hp) and is associated with its prognosis. We investigated the effect of the combination of HMGB1 and Hp on sepsis prognosis. METHODS: This single-center, retrospective study registered 78 patients with sepsis according to Sepsis-3 criteria on day 1 of diagnosis from July 2016 to November 2018. We divided the patients into four groups according to the serum concentration of 6.2 ng/mL HMGB1 and the median value of Hp. The 180-day mortality rates and cytokine concentrations of the low and high HMGB1 groups were compared. RESULTS: There was no difference in the 180-day mortality rate between the low Hp group and the high Hp group in the low HMGB1 group (P = 0.691). In the high HMGB1 group, a statistically significant difference was found between the low Hp group and the high Hp group (P = 0.002). In the high HMGB1 group, high Hp was associated with a better prognosis in univariate analysis (odds ratio, 0.131; 95% confidence interval [CI], 0.027-0.629; P = 0.011), and multivariate analysis (adjusted odds ratio, 0.086; 95% CI, 0.013-0.582; P = 0.009). In addition, in the high HMGB1 group, interleukin-8 levels were significantly higher in the low Hp group than in the high Hp group (P = 0.004). CONCLUSION: Patients with sepsis-induced high serum HMGB1 levels and low serum Hp levels could have a poor long-term prognosis.

12.
Eur Radiol Exp ; 6(1): 7, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35132475

ABSTRACT

BACKGROUND: Malignant mesothelioma (MM) is an aggressive tumor, with a poor prognosis, usually unresectable due to late diagnosis, mainly treated with chemotherapy. BoxA, a truncated form of "high mobility group box 1" (HMGB1), acting as an HMGB1 antagonist, might exert a defensive action against MM. We investigated the potential of BoxA for MM treatment using experimental 40-MHz ultrasound and optical imaging (OI) in a murine model. METHODS: Murine MM cells infected with a lentiviral vector expressing the luciferase gene were injected into the peritoneum of 14 BALB/c mice (7 × 104 AB1-B/c-LUC cells). These mice were randomized to treatment with BoxA (n = 7) or phosphate-buffered saline (controls, n = 7). The experiment was repeated with 40 mice divided into two groups (n = 20 + 20) and treated as above to confirm the result and achieve greater statistical power. Tumor presence was investigated by experimental ultrasound and OI; suspected peritoneal masses underwent histopathology and immunohistochemistry examination. RESULTS: In the first experiment, none of the 7 controls survived beyond day 27, whereas 4/7 BoxA-treated mice (57.1%) survived up to day 70. In the second experiment, 6/20 controls (30.0%) and 16/20 BoxA-treated mice (80.0%) were still alive at day 34 (p = 0.004). In both experiments, histology confirmed the malignant nature of masses detected using experimental ultrasound and OI. CONCLUSION: In our preclinical experience on a murine model, BoxA seems to exert a protective role toward MM. Both experimental ultrasound and OI proved to be reliable techniques for detecting MM peritoneal masses.


Subject(s)
HMGB1 Protein , Mesothelioma, Malignant , Animals , Disease Models, Animal , Mice , Optical Imaging , Ultrasonography
13.
Braz J Otorhinolaryngol ; 88(6): 875-881, 2022.
Article in English | MEDLINE | ID: mdl-33461910

ABSTRACT

INTRODUCTION: Serum level of high-mobility group box 1 protein is reportedly correlated with the severity of obstructive sleep apnea. OBJECTIVE: We tried to evaluate the possibility of using the serum high-mobility group box 1 protein level as a biologic marker in obstructive sleep apnea patients. METHODS: We generated a chronic intermittent hypoxia murine model that reflected human obstructive sleep apnea. Obstructive sleep apnea patients who underwent polysomnography were prospectively enrolled. Serum samples were obtained from mice and obstructive sleep apnea patients, and the serum high-mobility group box1 protein level was measured by enzyme-linked immunosorbent assay. RESULTS: Serum high-mobility group box 1 protein level was 56.16 ±â€¯30.33 ng/mL in chronic intermittent hypoxia and 18.63 ±â€¯6.20 ng/mL in control mice (p < 0.05). The mean apnea-hypopnea index and respiratory disturbance index values of enrolled obstructive sleep apnea patients were 50.35 ±â€¯27.96 and 51.56 ±â€¯28.53, respectively, and the mean serum high-mobility group box 1 protein level was 30.13 ±â€¯19.97 ng/mL. The apnea-hypopnea index and respiratory disturbance index were not significantly correlated with the serum high-mobility group box 1 protein level (p > 0.05). Instead, this protein level was significantly correlated with lowest arterial oxygen concentration (SaO2) (p < 0.05). CONCLUSION: High-mobility group box 1 protein may be involved in the pathogenesis of obstructive sleep apnea, and the possibility of this protein being a useful biologic marker in obstructive sleep apnea should be further evaluated.


Subject(s)
Sleep Apnea, Obstructive , Humans , Mice , Animals , Polysomnography , Hypoxia , Biomarkers
14.
Clin Res Hepatol Gastroenterol ; 46(2): 101791, 2022 02.
Article in English | MEDLINE | ID: mdl-34400366

ABSTRACT

BACKGROUND: Activation of the receptor for advanced glycation end products (RAGE) and its ligand High Mobility Group Box Protein 1 (HMGB1), a nuclear protein with proinflammatory properties, has been implicated in several inflammatory disorders. OBJECTIVE: To analyse the influence of RAGE and HMGB1 signalling in patients with primary sclerosing cholangitis (PSC). METHODS: Levels of HMGB1 and bile acid in serum and bile samples of 69 PSC patients and 32 controls were measured. Additionally, 640 patients with PSC and other liver diseases were analysed for the gain-of-function RAGE G82S SNP by PCR. Laboratory and clinical parameters were retrieved by chart review. RESULTS: ELISA analysis showed significantly higher biliary HMGB1 concentrations in PSC patients (n=69, median 124,1 ng/ml) than in the control group (n=32, median 6,85 ng/ml, p<0,001). Median serum HMGB1 (n=22, median 2,4 ng/ml) was significantly lower than median biliary HMGB1 of the concomitant bile samples (n=22, median 151 ng/ml, p =0,001). There was no correlation of biliary HMGB1 levels with laboratory parameters or clinical end points. Analysis of the gain-of-function G82SSNP RAGE SNP in PSC patients showed 8 patients with heterozygote mutant alleles (8/324, 2,5%). Patients carrying the mutation developed more often dominant strictures of the large bile ducts (100.0% vs. 61.3%; p=0.04) and had reduced transplantation-free survival in the mutant allele group (p<0.001). CONCLUSIONS: Biliary HMGB1 levels are elevated in PSC patients compared to controls and a gain-of-function SNP in RAGE is associated with development of dominant strictures and reduced survival in PSC patients.


Subject(s)
Biliary Tract , Cholangitis, Sclerosing , HMGB1 Protein , Receptor for Advanced Glycation End Products , Cholangitis, Sclerosing/genetics , Constriction, Pathologic , HMGB1 Protein/metabolism , Humans , Receptor for Advanced Glycation End Products/metabolism
15.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-930049

ABSTRACT

Tumor immunogenic cell death is a type of regulatory cell death, which is driven by stress including chemotherapy drugs, radiotherapy, oncolytic virus, nano carrier drugs and photodynamic force. It can induce specific immune response to tumor death cell antigen. The further study can provide theoretical basis and new ideas for anti-tumor immunity and clinical immunotherapy of tumor.

16.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-958121

ABSTRACT

Inflammation plays an important part in neonatal hypoxic-ischemic brain damage (HIBD). High mobility group box-1 protein (HMGB1), a neuroinflammatory trigger, has a dual effect on HIBD: in the acute stage, it amplifies the ischemic tissue injury; in the later stage, it is involved in the neurovascular repair and reconstruction. The significance of HMGB1 in the pathogenesis of HIBD is still not fully understood. This review summarizes the role of HMGB1 in HIBD, including its effects on neurons, glial cells and blood-brain barrier, and the underlying mechanisms as well as the progress in research on HMGB1 in immature brain, hoping to provide new ideas for neuroprotection in HIBD.

17.
Iran J Parasitol ; 16(3): 366-376, 2021.
Article in English | MEDLINE | ID: mdl-34630581

ABSTRACT

BACKGROUND: We aimed to determine whether neutralizing high mobility group box-1 (HMGB-1) prevents the release of HMGB-1 and proinflammatory cytokines on hemozoin (Hz)-induced alveolar epithelial cell in a model of malaria associated ALI/ARDS. METHODS: This study was conducted in the Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand in 2020. Human pulmonary alveolar epithelial cells (HPAEpiCs) were exposed to medium alone or 20 µM Hz for 24 h and incubated with different concentrations (1, 5, and 10 µg/ml) of anti-HMGB-1 monoclonal antibody (mAb) for various times (0, 4, 12, 24, and 48 h). The levels of HMGB-1, TNF-α and IFN-γ in the supernatants were measured by ELISA. The mRNA expression of RAGE, TLR-2 and TLR-4 were analyzed by real-time PCR. RESULTS: The HPAEpiCs treated with 10 µg/ml anti-HMGB-1 mAb showed a significant reduction in HMGB-1 release into the supernatant compared with those treated with 1 and 5 µg/ml anti-HMGB-1 mAb. The levels of TNF-α and IFN-γ were significantly decreased in the supernatant of HPAEpiCs treated with 1, 5, and 10 µg/ml anti-HMGB-1 mAb for 4, 12, 24, and 48 h compared with those stimulated with Hz alone. The mRNA expression levels of RAGE, TLR-2, and TLR-4 were significantly decreased after 24 h of anti-HMGB-1 antibody treatment at all concentrations. CONCLUSION: An anti-HMGB-1 antibody could be an effective agent for inhibiting the release of HMGB-1, TNF-α and IFN-γ. Furthermore, a neutralizing anti-HMGB-1 antibody could be applicable for the treatment of malaria-associated ALI/ARDS.

18.
Heart Lung ; 50(6): 885-892, 2021.
Article in English | MEDLINE | ID: mdl-34411870

ABSTRACT

BACKGROUND: The role of blood high mobility group box-1 (HMGB-1) protein in predicting mortality of sepsis remains controversial. OBJECTIVE: Here we conducted a meta-analysis to seek evidence for the association between blood HMGB-1 concentrations and mortality in patients with sepsis. METHODS: Eligible studies were identified by a comprehensive search of six digital databases, supplemented by a manual search of related references. Standardized mean differences (SMDs) and corresponding 95% confidence intervals (CIs) were calculated as effect estimates. RESULTS: A total of eighteen studies, covering 1163 patients with sepsis, were included. Compared with survival groups of sepsis, non-survival groups had significantly higher blood HMGB-1 concentrations at enrollment (SMD: 0.45, 95% CI: 0.21-0.69). Subgroup analyses showed that no significant differences were found between two groups among patients with more severe sepsis (SMD: 0.18, 95% CI: -0.02-0.38). A significant association between initial HMGB-1 levels and ≤30-day mortality remained (SMD: 0.43, 95% CI: 0.09-0.78). Besides, HMGB-1 levels were observed to be more significantly higher in non-survival groups after the third day of admission (SMD: 1.33, 95% CI: 1.05-1.62) but two groups attained comparable HMGB-1 levels on day 7 (SMD: 1.01, 95% CI: -0.31-2.33). CONCLUSIONS: Initial high blood HMGB-1 levels are significantly associated with short-term (≤30 days) mortality of patients with sepsis, and the association may be affected by the severity of sepsis. Subsequent monitoring of HMGB-1 levels, on the third and seventh day after admission, is encouraged for better evaluation of HMGB-1 as a prognostic marker of mortality in sepsis.


Subject(s)
Sepsis , Hospitalization , Humans
19.
J Cell Mol Med ; 25(18): 8947-8956, 2021 09.
Article in English | MEDLINE | ID: mdl-34405526

ABSTRACT

Amyloid-ß (Aß) deposition in the brain has been implicated in the development of Alzheimer's disease (AD), and neuroinflammation generates AD progression. Therapeutic effects of anti-inflammatory approaches in AD are still under investigation. Curcumin, a potent anti-inflammatory and antioxidant, has demonstrated therapeutic potential in AD models. However, curcumin's anti-inflammatory molecular mechanisms and its associated cognitive impairment mechanisms in AD remain unclear. The high-mobility group box-1 protein (HMGB1) participates in the regulation of neuroinflammation. Herein, we attempted to evaluate the anti-inflammatory effects of chronic oral administration of curcumin and HMGB1 expression in APP/PS1 transgenic mice AD model. We found that transgenic mice treated with a curcumin diet had shorter escape latencies and showed a significant increase in percent alternation, when compared with transgenic mice, in the Morris water maze and Y-maze tests. Additionally, curcumin treatment could effectively decrease HMGB1 protein expression, advanced glycosylation end product-specific receptor (RAGE), Toll-like receptors-4 (TLR4) and nuclear factor kappa B (NF-κB) in transgenic mice hippocampus. However, amyloid plaques detected with thioflavin-S staining in transgenic mice hippocampus were not affected by curcumin treatment. In contrast, curcumin significantly decreased GFAP-positive cells, as assessed by immunofluorescence staining. Taken together, these data indicate that oral administration of curcumin may be a promising agent to attenuate memory deterioration in AD mice, probably inhibiting the HMGB1-RAGE/TLR4-NF-κB inflammatory signalling pathway.


Subject(s)
Alzheimer Disease/drug therapy , Curcumin/pharmacology , HMGB1 Protein/metabolism , Hippocampus/drug effects , Animals , Hippocampus/pathology , Male , Mice , Mice, Transgenic , Receptor for Advanced Glycation End Products/metabolism
20.
Pediatr Rheumatol Online J ; 19(1): 112, 2021 Jul 12.
Article in English | MEDLINE | ID: mdl-34247641

ABSTRACT

OBJECTIVE: To analyze the levels of high mobility group box 1 (HMGB1) protein on different courses of juvenile idiopathic arthritis (JIA). METHODS: In our prospective longitudinal study, children with JIA were included with their blood samples collected at the first visit, 1-month, 3-month, and 6-month follow-up, respectively. Samples were also collected from healthy controls and children with reactive arthritis at the first visit. Levels of HMGB1 were determined using enzyme-linked immunosorbent assays. Clinical disease characteristics and routine laboratory findings were analyzed as well. RESULTS: A total of 64 children were enrolled, of whom 31 (48.4%) were female. The median age at the first visit for participants with JIA was 9.25 years (range, 1.42-15.42) and the median duration of disease was 2.38 months (range, 1.53-49.31). Serum HMGB1 levels at the first visit were significantly elevated in children with systemic JIA compared with other groups, and so were in enthesitis-related arthritis versus healthy controls. Significant correlations were established at the first visit between HMGB1 levels and duration of disease, C-reactive protein, percentage of neutrophils, and ferritin. Data from all samples revealed that serum HMGB1 levels in JIA were significantly associated with erythrocyte sedimentation rates, C-reactive protein, percentage of neutrophils, and disease activity scores. CONCLUSIONS: Serum HMGB1 may be associated with clinical disease activity of JIA and specifically increased at the first visit in children with systemic JIA, suggesting its function as a sensitive inflammatory marker. Further large-scale studies are warranted to explore its spectrum in JIA.


Subject(s)
Arthritis, Juvenile/blood , HMGB1 Protein/blood , Adolescent , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Infant , Longitudinal Studies , Male , Prospective Studies , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL