Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 505
Filter
1.
Drug Dev Res ; 85(4): e22219, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38845211

ABSTRACT

Sepsis is a life-threatening organ dysfunction that endangers patient lives and is caused by an imbalance in the host defense against infection. Sepsis continues to be a significant cause of morbidity and mortality in critically sick patients. Oxymatrine (OMT), a quinolizidine alkaloid derived from the traditional Chinese herb Sophora flavescens Aiton, has been shown to have anti-inflammatory effects on a number of inflammatory illnesses according to research. In this study, we aimed to evaluate the therapeutic effects of OMT on sepsis and explore the underlying mechanisms. We differentiated THP-1 cells into THP-1 macrophages and studied the anti-inflammatory mechanism of OMT in a lipopolysaccharide (LPS)-induced THP-1 macrophage sepsis model. Activation of the receptor for advanced glycation end products (RAGE), as well as NF-κB, was assessed by Western blot analysis and immunofluorescence staining. ELISA was used to measure the levels of inflammatory factors. We found that OMT significantly inhibited HMGB1-mediated RAGE/NF-κB activation and downstream inflammatory cytokine production in response to LPS stimulation. Finally, an in vivo experiment was performed on septic mice to further study the effect of OMT on injured organs. The animal experiments showed that OMT significantly inhibited HMGB1-mediated RAGE/NF-κB activation, protected against the inflammatory response and organ injury induced by CLP, and prolonged the survival rate of septic mice. Herein, we provide evidence that OMT exerts a significant therapeutic effect on sepsis by inhibiting the HMGB1/RAGE/NF-κB signaling pathway.


Subject(s)
Alkaloids , HMGB1 Protein , Inflammation , Lipopolysaccharides , NF-kappa B , Quinolizines , Receptor for Advanced Glycation End Products , Sepsis , Signal Transduction , Alkaloids/pharmacology , Alkaloids/therapeutic use , Quinolizines/pharmacology , Quinolizines/therapeutic use , Animals , Sepsis/drug therapy , Sepsis/complications , Sepsis/metabolism , NF-kappa B/metabolism , HMGB1 Protein/metabolism , HMGB1 Protein/antagonists & inhibitors , Humans , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction/drug effects , Mice , Inflammation/drug therapy , Inflammation/metabolism , Male , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , THP-1 Cells , Mice, Inbred C57BL , Macrophages/drug effects , Macrophages/metabolism , Matrines
2.
J Neuroimmune Pharmacol ; 19(1): 21, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38771510

ABSTRACT

The neurotoxicity of Semen Strychni has been reported recently in several clinical cases. Therefore, this study was conducted to investigate the role of HMGB1 in a model of neurotoxicity induced by Semen Strychni and to assess the potential alleviating effects of glycyrrhizic acid (GA), which is associated with the regulation of HMGB1 release. Forty-eight SD rats were intraperitoneally injected with Semen Strychni extract (175 mg/kg), followed by oral administration of GA (50 mg/kg) for four days. After treatment of SS and GA, neuronal degeneration, apoptosis, and necrosis were observed via histopathological examination. Inflammatory cytokines (TNF-α and IL-1ß), neurotransmitter associated enzymes (MAO and AChE), serum HMGB1, nuclear and cytoplasmic HMGB1/ph-HMGB1, and the interaction between PP2A, PKC, and HMGB1 were evaluated. The influence of the MAPK pathway was also examined. As a result, this neurotoxicity was characterized by neuronal degeneration and apoptosis, the induction of pro-inflammatory cytokines, and a reduction in neurotransmitter-metabolizing enzymes. In contrast, GA treatment significantly ameliorated the abovementioned effects and alleviated nerve injury. Furthermore, Semen Strychni promoted HMGB1 phosphorylation and its translocation between the nucleus and cytoplasm, thereby activating the NF-κB and MAPK pathways, initiating various inflammatory responses. Our experiments demonstrated that GA could partially reverse these effects. In summary, GA acid alleviated Semen Strychni-induced neurotoxicity, possibly by inhibiting HMGB1 phosphorylation and preventing its release from the cell.


Subject(s)
Glycyrrhizic Acid , HMGB1 Protein , Rats, Sprague-Dawley , Animals , Glycyrrhizic Acid/pharmacology , Glycyrrhizic Acid/therapeutic use , HMGB1 Protein/metabolism , HMGB1 Protein/antagonists & inhibitors , Rats , Male , Phosphorylation/drug effects , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/metabolism
3.
Expert Opin Ther Pat ; 34(3): 127-140, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38557201

ABSTRACT

INTRODUCTION: HMGB1 is a non-histone chromatin protein released or secreted in response to tissue damage or infection. Extracellular HMGB1, as a crucial immunomodulatory factor, binds with several different receptors to innate inflammatory responses that aggravate acute and chronic liver diseases. The increased levels of HMGB1 have been reported in various liver diseases, highlighting that it represents a potential biomarker and druggable target for therapeutic development. AREAS COVERED: This review summarizes the current knowledge on the structure, function, and interacting receptors of HMGB1 and its significance in multiple liver diseases. The latest patented and preclinical studies of HMGB1 inhibitors (antibodies, peptides, and small molecules) for liver diseases are summarized by using the keywords 'HMGB1,' 'HMGB1 antagonist, HMGB1-inhibitor,' 'liver disease' in Web of Science, Google Scholar, Google Patents, and PubMed databases in the year from 2017 to 2023. EXPERT OPINIONS: In recent years, extensive research on HMGB1-dependent inflammatory signaling has discovered potent inhibitors of HMGB1 to reduce the severity of liver injury. Despite significant progress in the development of HMGB1 antagonists, few of them are approved for clinical treatment of liver-related diseases. Developing safe and effective specific inhibitors for different HMGB1 isoforms and their interaction with receptors is the focus of future research.


Subject(s)
Drug Development , HMGB1 Protein , Liver Diseases , Patents as Topic , Humans , HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/metabolism , Liver Diseases/drug therapy , Liver Diseases/physiopathology , Animals , Biomarkers/metabolism , Inflammation/drug therapy , Inflammation/physiopathology , Severity of Illness Index
4.
Ecotoxicol Environ Saf ; 266: 115575, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37839183

ABSTRACT

Exposure to toluene diisocyanate (TDI) can cause pulmonary diseases such as asthma. Inhibition of high mobility group box 1 protein (HMGB1) has been found to be protective against the toxic effects of TDI on human bronchial epithelial (HBE) cells. Here, we evaluated the in vivo positive roles of HMGB1 in the TDI-caused asthma mice and explored its underlying mechanisms in HBE cells. We found that suppression of HMGB1 obviously alleviated airway inflammation, airway hyperresponsiveness, and airway remodeling in the lung tissue of the asthma mice. The in vitro results showed that inhibition of HMGB1 ameliorated TDI-induced reactive oxygen species (ROS) release, inflammatory response, and activation of autophagy in HBE cells. At the molecular level, inhibition of HMGB1 decreased the expressions of HMGB1, Toll-like receptor 4, Vimentin and matrix metalloproteinase-9 proteins, activated NF-κB and NOD-like receptor protein 3 (NLRP3) inflammasome, and increased E-cadherin expression. Importantly, activation of autophagy could lead to the overactivation of NLRP3 inflammasome in TDI-induced asthma. These results suggest that inhibition of HMGB1 can alleviate TDI-induced asthma through ROS/AMPK/autophagy pathways, which may provide valuable evidence for the pathogenesis and therapeutic targets of TDI-induced asthma.


Subject(s)
Asthma, Occupational , HMGB1 Protein , Toluene 2,4-Diisocyanate , Animals , Humans , Mice , AMP-Activated Protein Kinases/metabolism , Asthma, Occupational/drug therapy , Asthma, Occupational/pathology , HMGB1 Protein/antagonists & inhibitors , Inflammasomes/metabolism , Lung , Mice, Inbred BALB C , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reactive Oxygen Species/metabolism , Toluene 2,4-Diisocyanate/pharmacology , Toluene 2,4-Diisocyanate/toxicity
5.
Immunology ; 169(2): 117-131, 2023 06.
Article in English | MEDLINE | ID: mdl-36571562

ABSTRACT

Hyperinflammation characterized by elevated proinflammatory cytokines known as 'cytokine storms' is the major cause of high severity and mortality seen in COVID-19 patients. The pathology behind the cytokine storms is currently unknown. Increased HMGB1 levels in serum/plasma of COVID-19 patients were reported by many studies, which positively correlated with the level of proinflammatory cytokines. Dead cells following SARS-CoV-2 infection might release a large amount of HMGB1 and RNA of SARS-CoV-2 into extracellular space. HMGB1 is a well-known inflammatory mediator. Additionally, extracellular HMGB1 might interact with SARS-CoV-2 RNA because of its high capability to bind with a wide variety of molecules including nucleic acids and could trigger massive proinflammatory immune responses. This review aimed to critically explore the many possible pathways by which HMGB1-SARS-CoV-2 RNA complexes mediate proinflammatory responses in COVID-19. The contribution of these pathways to impair host immune responses against SARS-CoV-2 infection leading to a cytokine storm was also evaluated. Moreover, since blocking the HMGB1-SARS-CoV-2 RNA interaction might have therapeutic value, some of the HMGB1 antagonists have been reviewed. The HMGB1- SARS-CoV-2 RNA complexes might trigger endocytosis via RAGE which is linked to lysosomal rupture, PRRs activation, and pyroptotic death. High levels of the proinflammatory cytokines produced might suppress many immune cells leading to uncontrolled viral infection and cell damage with more HMGB1 released. Altogether these mechanisms might initiate a proinflammatory cycle leading to a cytokine storm. HMGB1 antagonists could be considered to give benefit in alleviating cytokine storms and serve as a potential candidate for COVID-19 therapy.


Subject(s)
Antiviral Agents , COVID-19 Drug Treatment , COVID-19 , Cytokine Release Syndrome , HMGB1 Protein , Molecular Targeted Therapy , RNA, Viral , SARS-CoV-2 , Humans , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/virology , COVID-19/complications , COVID-19/immunology , HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/metabolism , RNA, Viral/metabolism , Host Microbial Interactions/immunology , SARS-CoV-2/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use
6.
Toxicol Lett ; 373: 22-32, 2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36375637

ABSTRACT

Dasatinib, a second-generation BCR-ABL inhibitor, is currently used as first-line treatment for patients with chronic myeloid leukemia. However, dasatinib treatment increases the risk of severe cutaneous toxicity, which limits its long-term safe use in clinic. The underlying mechanism for dasatinib-induced cutaneous toxicity has not been clarified. In this study, we tested the toxicity of dasatinib on human immortal keratinocyte line (HaCaT) and normal human epidermal keratinocytes (NHEK). We found that dasatinib directly caused cytotoxicity on keratinocytes, which could be the explanation of the clinical characteristic of pathology. Mechanistically, dasatinib impaired mitophagy by downregulating HMGB1 protein level in keratinocytes, which led to the accumulation of dysfunctional mitochondria. Mitochondria-derived ROS caused DNA damage and cell apoptosis. More importantly, we confirmed that overexpression of HMGB1 could reverse dasatinib-induced keratinocyte apoptosis, and preliminarily explored the intervention effect of saikosaponin A, which could increase HMGB1 expression, on cutaneous toxicity caused by dasatinib. Collectively, our study revealed that dasatinib induced keratinocyte apoptosis via inhibiting HMGB1-mediated mitophagy and saikosaponin A could be a viable strategy for prevention of dasatinib-induced cutaneous toxicity.


Subject(s)
Apoptosis , Dasatinib , Humans , Apoptosis/drug effects , Dasatinib/toxicity , HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/drug effects , HMGB1 Protein/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Mitophagy/drug effects , Protein Kinase Inhibitors/pharmacology
7.
Mol Med ; 28(1): 108, 2022 09 07.
Article in English | MEDLINE | ID: mdl-36071400

ABSTRACT

BACKGROUND: High-mobility group box 1 protein (HMGB1) is an ubiquitous nuclear protein that once released in the extracellular space acts as a Damage Associated Molecular Pattern and promotes inflammation. HMGB1 is significantly elevated during Pseudomonas aeruginosa infections and has a clinical relevance in respiratory diseases such as Cystic Fibrosis (CF). Salicylates are HMGB1 inhibitors. To address pharmacological inhibition of HMGB1 with small molecules, we explored the therapeutic potential of pamoic acid (PAM), a salicylate with limited ability to cross epithelial barriers. METHODS: PAM binding to HMGB1 and CXCL12 was tested by Nuclear Magnetic Resonance Spectroscopy using chemical shift perturbation methods, and inhibition of HMGB1·CXCL12-dependent chemotaxis was investigated by cell migration experiments. Aerosol delivery of PAM, with single or repeated administrations, was tested in murine models of acute and chronic P. aeruginosa pulmonary infection in C57Bl/6NCrlBR mice. PAM efficacy was evaluated by read-outs including weight loss, bacterial load and inflammatory response in lung and bronco-alveolar lavage fluid. RESULTS: Our data and three-dimensional models show that PAM is a direct ligand of both HMGB1 and CXCL12. We also showed that PAM is able to interfere with heterocomplex formation and the related chemotaxis in vitro. Importantly, PAM treatment by aerosol was effective in reducing acute and chronic airway murine inflammation and damage induced by P. aeruginosa. The results indicated that PAM reduces leukocyte recruitment in the airways, in particular neutrophils, suggesting an impaired in vivo chemotaxis. This was associated with decreased myeloperoxidase and neutrophil elastase levels. Modestly increased bacterial burdens were recorded with single administration of PAM in acute infection; however, repeated administration in chronic infection did not affect bacterial burdens, indicating that the interference of PAM with the immune system has a limited risk of pulmonary exacerbation. CONCLUSIONS: This work established the efficacy of treating inflammation in chronic respiratory diseases, including bacterial infections, by topical delivery in the lung of PAM, an inhibitor of HMGB1.


Subject(s)
Chemokine CXCL12 , HMGB1 Protein , Naphthols , Pneumonia, Bacterial , Animals , Chemokine CXCL12/antagonists & inhibitors , Chemotaxis/drug effects , Disease Models, Animal , HMGB1 Protein/antagonists & inhibitors , Inflammation/drug therapy , Inflammation/pathology , Mice , Mice, Inbred C57BL , Naphthols/pharmacology , Pneumonia, Bacterial/drug therapy , Pseudomonas aeruginosa/metabolism
8.
Nat Commun ; 13(1): 166, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013338

ABSTRACT

Muscle cell death in polymyositis is induced by CD8+ cytotoxic T lymphocytes. We hypothesized that the injured muscle fibers release pro-inflammatory molecules, which would further accelerate CD8+ cytotoxic T lymphocytes-induced muscle injury, and inhibition of the cell death of muscle fibers could be a novel therapeutic strategy to suppress both muscle injury and inflammation in polymyositis. Here, we show that the pattern of cell death of muscle fibers in polymyositis is FAS ligand-dependent necroptosis, while that of satellite cells and myoblasts is perforin 1/granzyme B-dependent apoptosis, using human muscle biopsy specimens of polymyositis patients and models of polymyositis in vitro and in vivo. Inhibition of necroptosis suppresses not only CD8+ cytotoxic T lymphocytes-induced cell death of myotubes but also the release of inflammatory molecules including HMGB1. Treatment with a necroptosis inhibitor or anti-HMGB1 antibodies ameliorates myositis-induced muscle weakness as well as muscle cell death and inflammation in the muscles. Thus, targeting necroptosis in muscle cells is a promising strategy for treating polymyositis providing an alternative to current therapies directed at leukocytes.


Subject(s)
HMGB1 Protein/antagonists & inhibitors , Imidazoles/pharmacology , Indoles/pharmacology , Muscle Fibers, Skeletal/drug effects , Myositis/prevention & control , Necroptosis/drug effects , Polymyositis/genetics , Animals , Antibodies, Neutralizing/pharmacology , C-Reactive Protein/administration & dosage , Fas Ligand Protein/genetics , Fas Ligand Protein/immunology , Female , Gene Expression Regulation , Granzymes/genetics , Granzymes/immunology , HMGB1 Protein/genetics , HMGB1 Protein/immunology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Fibers, Skeletal/immunology , Muscle Fibers, Skeletal/pathology , Muscle Strength/drug effects , Muscle Strength/immunology , Muscle, Skeletal/drug effects , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Myositis/chemically induced , Myositis/genetics , Myositis/immunology , Necroptosis/genetics , Necroptosis/immunology , Perforin/genetics , Perforin/immunology , Polymyositis/immunology , Polymyositis/pathology , Signal Transduction , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology
9.
Bioorg Med Chem Lett ; 58: 128523, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34973341

ABSTRACT

For the discovery of new pentacyclic triterpenes as a potential anti-inflammatory agent, microbial transformation of uvaol by Penicilium griseofulvum CICC 40293 and Streptomyces griseus ATCC 13273 was investigated. Stereoselective hydroxylation and epoxidation reactions were observed in the biotransformation. Moreover, six new metabolites were isolated and structurally elucidated by HR-ESI-MS and NMR spectrum. All the compounds were evaluated upon the inhibitory effects of nitric oxide (NO) release in RAW 264.7 cells induced by lipopolysaccharide (LPS) and high-mobility group box 1 (HMGB1). Among them, compound 3 (13, 28-epoxy-3ß, 7ß, 21ß-trihydroxy-urs-11-ene) with the unique epoxy structure and compound 5 (3ß, 21ß, 24, 28-tetrahydroxy-urs-12-en-30-oic acid), exhibited a considerable inhibitory effect on both models while compound 2 (urs-12-ene-3ß, 7ß, 21ß, 28-tetraol) showed a significant bias in the LPS-induced inflammatory response with IC50 value of 2.22 µM. Therefore, this study could provide some insights on the discovery of the pentacyclic triterpene leads for the treatment of either DAMPs or PAMPs triggered inflammation.


Subject(s)
HMGB1 Protein/antagonists & inhibitors , Lipopolysaccharides/antagonists & inhibitors , Nitric Oxide/antagonists & inhibitors , Triterpenes/pharmacology , Animals , Dose-Response Relationship, Drug , HMGB1 Protein/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Molecular Structure , Nitric Oxide/biosynthesis , RAW 264.7 Cells , Structure-Activity Relationship , Triterpenes/chemical synthesis , Triterpenes/chemistry
10.
Inflammation ; 45(1): 172-179, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34480250

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection (COVID-19) is associated with uncontrolled inflammatory responses. Loss of pulmonary angiotensin-converting enzyme 2 (ACE2) function has been associated with SARS-CoV-2 infection. The aberrant signalling and dysregulated inflammation characteristic of lung cancer have marked similarities with SARS-CoV-2 infection. Spearman's correlation analysis of The Cancer Genome Atlas (TCGA) datasets indicated an inverse correlation between ACE2 and IL6 in lung adenocarcinoma. qRT-PCR analysis revealed CoV-2-SRBD-mediated diminished ACE2 expression in lung cancer cells that was concomitant with increased IL6 expression. Western blot and qRT-PCR analysis suggested that treatment with methotrexate (MTx) dampened CoV-2-SRBD-mediated increase in JAK1/STAT3 phosphorylation, gp130, IL6, and folate-binding protein (FBP) expressions. MTx also rescued the diminished expression of ACE2 in CoV-2-SRBD transfected cells. As lung tissue injury in severely affected COVID-19 patients is characterised by aberrant inflammatory response, repurposing MTx as an effective therapy against critical regulators of inflammation in SARS-CoV-2 infection warrants investigation.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19 Drug Treatment , Glycyrrhizic Acid/therapeutic use , Immunosuppressive Agents/therapeutic use , Interleukin-6/biosynthesis , Methotrexate/therapeutic use , A549 Cells , Adenocarcinoma of Lung/pathology , Anti-Inflammatory Agents/therapeutic use , COVID-19/immunology , COVID-19/pathology , Cell Line, Tumor , Cytokine Receptor gp130/biosynthesis , Folate Receptor 2/biosynthesis , HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/metabolism , Humans , Interleukin-6/immunology , Janus Kinase 1/metabolism , Lung Neoplasms/pathology , Phosphorylation/drug effects , SARS-CoV-2/drug effects , STAT3 Transcription Factor/metabolism , Spike Glycoprotein, Coronavirus/immunology
11.
Kaohsiung J Med Sci ; 38(3): 253-260, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34806822

ABSTRACT

Inflammation plays an important role during sepsis, and excessive inflammation can result in organ damage, chronic inflammation, fibrosis, and scarring. The study aimed to investigate the specific mechanism of emodin by constructing in vivo and in vitro septic lung injury models via inhibition and reduction of NF-kB and high mobility group box 1 (HMGB1) pathways. A cecal ligation and puncture (CLP) model was built for adult male Sprague-Dawley rats. Concentrations of TNF-α, IL-1ß, and IL-6 in bronchoalveolar lavage fluid were determined using commercially available ELISA kits. Hematoxylin and eosin staining was used for the right lung inferior lobes. Myeloperoxidase (MPO) activity of the lung tissue was detected by using the MPO kit. Murine alveolar epithelial cell line (MLE-12) cells were used for flow cytometry and Western blot to analyze the apoptosis rate and protein expression. Emodin significantly decreased CLP-induced cell apoptosis, upregulated expression of sirtuin 1 (SIRT1), and inhibited p-p65/p65 and HMGB1. In lipopolysaccharide (LPS) treated cell model, emodin treatment markedly decreased LPS-induced release of IL-1, IL-6, and tumor necrosis factor (TNF)-α, inhibited LPS-induced cell apoptosis and suppressed protein levels of P-P65/P65 and HMGB1. However, science of SIRT1 reversed the above effects by treatment of emodin. In summarize, this study found that emodin can alleviate sepsis-induced lung injury in vivo and in vitro through regulation of SIRT1.


Subject(s)
Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Emodin/therapeutic use , HMGB1 Protein/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Sepsis/complications , Sirtuin 1/metabolism , Animals , Bronchoalveolar Lavage , Cell Line , Emodin/pharmacology , HMGB1 Protein/metabolism , Interleukin-1beta/analysis , Interleukin-6/analysis , Male , Mice , NF-kappa B/metabolism , Peroxidase/metabolism , Rats, Sprague-Dawley , Signal Transduction , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/analysis
12.
Exp Parasitol ; 231: 108171, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34736899

ABSTRACT

Schistosomiasis is a prevalent zoonotic parasitic disease caused by schistosomes. Its main threat to human health is hepatic granuloma and fibrosis due to worm eggs. Praziquantel remains the first choice for the treatment of schistosomiasis but has limited benefit in treating liver fibrosis. Therefore, the need to develop effective drugs for treating schistosomiasis-induced hepatic fibrosis is urgent. High-mobility group box 1 protein (HMGB1) is a potential immune mediator that is highly associated with the development of some fibrotic diseases and may be involved in the liver pathology of schistosomiasis. We speculated that HMGB1 inhibitors could have an anti-fibrotic effect. Sodium butyrate (SB), a potent inhibitor of HMGB1, has shown anti-inflammatory activity in some animal disease models. In this study, we evaluated the effects of SB on a murine schistosomiasis model. Mice were percutaneously infected with 20 ± 2 cercariae of Schistosoma japonicum. SB (500 mg/kg/day) was administered every 3 days for the entire experiment period. The activity of alanine aminotransferase (ALT) and aspartate aminotransferase (AST), liver histopathology, HMGB1 expression, and the levels of interferon gamma (IFN-γ), transforming growth factor-ß1 (TGF-ß1), and interleukin-6 (IL-6) in serum were analyzed. SB reduced hepatic granuloma and fibrosis of schistosomiasis, reflected by the decreased levels of ALT and AST in serum and the reduced expression of pro-inflammatory and fibrogenic cytokines (IFN-γ, TGF-ß1, and IL-6). The protective effect could be attributable to the inhibition of the expression of HMGB1 and release by SB.


Subject(s)
Butyric Acid/pharmacology , Butyric Acid/therapeutic use , HMGB1 Protein/antagonists & inhibitors , Liver Cirrhosis/drug therapy , Schistosoma japonicum/drug effects , Schistosomiasis japonica/drug therapy , Alanine Transaminase/analysis , Animals , Aspartate Aminotransferases/analysis , Blotting, Western , Cytokines/blood , Disease Models, Animal , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Female , HMGB1 Protein/genetics , Histamine Antagonists/pharmacology , Histamine Antagonists/therapeutic use , Humans , Liver/enzymology , Liver/metabolism , Liver/parasitology , Liver Cirrhosis/parasitology , Mice , Mice, Inbred C57BL , Neglected Diseases/drug therapy , Neglected Diseases/parasitology , Real-Time Polymerase Chain Reaction , Schistosomiasis japonica/complications , Schistosomiasis japonica/immunology , Specific Pathogen-Free Organisms , Zoonoses/parasitology
13.
Bioorg Med Chem ; 52: 116494, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34800877

ABSTRACT

High mobility group box-1 protein (HMGB1) is a typical Damage-Associated Molecular Patterns (DAMPs) released in response to cellular inflammation. The pentacyclic triterpenes (PTs) are considered to be the natural inhibitors against HMGB1-related inflammation. To explore new lead compounds of PTs as anti-inflammatory agents, biotransformation of four PTs by Streptomyces olivaceus CICC 23628 was investigated in this study. As a result, thirteen unique 3,4-seco-triterpenes metabolites were isolated and twelve of them were first identified and reported. Structures of metabolites were determined based on HR-ESI-MS, 1D/2D NMR, and single-crystal X-ray diffraction. Furthermore, all compounds were subjected to the bioassay on the model of HMGB1-stimulated RAW 264.7 cells to evaluate their anti-inflammatory activity through nitric oxide (NO) inhibition activity. Compounds 3b (3,4-seco-olean-12-en-4,21α,22ß,24-tetrahydroxy-ol-3-oic acid) and 2b (3,4-seco-olean-12-en-4,21ß,22ß,24,29-pentahydroxy-ol-3-oic acid) exhibited NO inhibitory activity with IC50 values of 15.94 µM and 36.00 µM, respectively. Thus, indicating their potential as HMGB1 inhibitors and in developing potent anti-inflammatory agents. This work provides an operationally simple, efficient method for the rapid diversification of the PTs scaffold for a variety of distinctive 3,4-seco-triterpenes to facilitate the discovery of potential anti-inflammatory compounds.


Subject(s)
Anti-Inflammatory Agents/pharmacology , HMGB1 Protein/antagonists & inhibitors , Pentacyclic Triterpenes/pharmacology , Streptomyces/metabolism , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/metabolism , Biotransformation , Cells, Cultured , Dose-Response Relationship, Drug , HMGB1 Protein/metabolism , Mice , Molecular Structure , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Pentacyclic Triterpenes/chemistry , Pentacyclic Triterpenes/metabolism , RAW 264.7 Cells , Structure-Activity Relationship
14.
Mol Med Rep ; 24(5)2021 Nov.
Article in English | MEDLINE | ID: mdl-34476498

ABSTRACT

Gycyrrhizic acid (GA), an inhibitor of high mobility group box 1 (HMGB1), inhibits inflammatory responses and is involved in the occurrence and development of several inflammation­related diseases. However, the role of GA in the atherosclerotic lesions caused by diabetes mellitus (DM) remains unknown. In the present study, Sprague Dawley rats were selected to desi=gn a diabetic atherosclerosis (AS) model. Rats from the DM­AS group were subsequently divided into DM­AS, DM­AS + GA (50 mg/kg) and DM­AS + GA (150 mg/kg) groups. Biochemical analyzers were used to measure levels of blood glucose, fasting insulin, total cholesterol, total triglyceride, low­density lipoprotein and high­density lipoprotein. The number of plaques was recorded after collection of thoracic aortas from the rats. The intimal thickness of arterial tissue was detected by hematoxylin and eosin staining. The expression levels of CD68 and α­smooth muscle actin (α­SMA) were detected by immunohistochemistry. The expression of tumor necrosis factor­α, interleukin (IL)­6 and IL­1ß in the serum of the rats was detected by ELISA. The expression of fatty acid synthetase, sterol regulatory element binding protein 1C, HMGB1 and receptor for advanced glycation end products (RAGE) was detected by western blotting. Reverse transcription quantitative PCR was used to detect the mRNA expression of HMGB1 and RAGE. The results demonstrated that GA treatment could decrease the body weight, blood glucose level and biochemical parameters of AS DM rats in a dose­dependent manner. In addition, GA decreased the intimal thickness of carotid artery and the formation of plaque in rats with diabetic AS. Furthermore, GA inhibited macrophage activation and decreased α­SMA expression in vascular smooth muscle cells, and decreased the expression of proteins (FAS and SREBP­1c) and inflammatory factors. Taken together, the findings from the present study demonstrated that GA may have a therapeutic effect on DM­associated AS. This study provides a theoretical basis for the treatment of diabetic AS.


Subject(s)
Atherosclerosis/drug therapy , Diabetes Mellitus, Experimental/complications , Glycyrrhizic Acid/pharmacology , HMGB1 Protein/antagonists & inhibitors , Animals , Atherosclerosis/etiology , Diabetes Mellitus, Experimental/chemically induced , Glycyrrhizic Acid/therapeutic use , HMGB1 Protein/metabolism , Humans , Macrophage Activation/drug effects , Male , Rats , Rats, Sprague-Dawley , Streptozocin/administration & dosage , Streptozocin/toxicity
15.
Exp Cell Res ; 409(2): 112842, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34563514

ABSTRACT

We explore the role of miR-125b in septic cardiomyopathy, focusing on miR-125b/STAT3/HMGB1 axis. CLP mouse model and LPS-stimulated primary rat cardiomyocytes (CMs) and H9C2 cell were used as in vivo and in vitro models of septic cardiomyopathy, respectively. qRT-PCR and western blot were performed to measure expression levels of miR-125b, STAT3, HMGB1, and autophagy-related proteins. MTT assay was employed to examine LPS toxicity. Dual luciferase activity assay and CHIP were performed to validate interactions of miR-125b/STAT3 and STAT3/HMGB1 promoter. Immunostaining was used to assess the level of autophagic flux. ROS level was measured by fluorescence assay. Heart functions were examined via intracoronary Doppler ultrasound. miR-125b was diminished while STAT3 and HMGB1 were elevated in the heart tissue following CLP surgery and in LPS-treated H9C2 cells. LPS treatment up-regulated ROS generation and suppressed autophagic flux. Overexpression of miR-125b mimics or knockdown of STAT3 or HMGB1 alleviated LPS-induced hindrance of autophagic flux and ROS production. miR-125b directly targeted STAT3 mRNA and STAT3 bound with HMGB1 promoter. Overexpression of miR-125b mitigated myocardial dysfunction induced by CLP in vivo. Hyperactivation of STAT3/HMGB1 caused by reduced miR-125b contributes to ROS generation and the hindrance of autophagic flux during septic cardiomyopathy, leading to myocardial dysfunction.


Subject(s)
Autophagy , Cardiomyopathies/prevention & control , HMGB1 Protein/antagonists & inhibitors , MicroRNAs/genetics , STAT3 Transcription Factor/antagonists & inhibitors , Sepsis/complications , Animals , Apoptosis , Cardiomyopathies/etiology , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cell Proliferation , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Mice , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction
16.
Sci Rep ; 11(1): 18356, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34526618

ABSTRACT

Glycyrrhizin (GL) is a direct inhibitor of HMGB1 which acts as an alarmin when excreted into the extracellular space. High-dose radiation in radiotherapy induces collateral damage to the normal tissue, which can be mitigated by GL inhibiting HMGB1. The purpose of this study was to assess changes in HMGB1 and pro-inflammatory cytokines and to evaluate the protective effect of GL after low-dose radiation exposure. BALB/c mice were irradiated with 0.1 Gy (n = 10) and 1 Gy (n = 10) with GL being administered to half of the mice (n = 5, respectively) before irradiation. Blood and spleen samples were harvested and assessed for oxidative stress, HMGB1, pro-inflammatory cytokines, and cell viability. HMGB1 and pro-inflammatory cytokines increased and cell viability decreased after irradiation in a dose-dependent manner. Oxidative stress also increased after irradiation, but did not differ between 0.1 Gy and 1 Gy. With the pretreatment of GL, oxidative stress, HMGB1, and all of the pro-inflammatory cytokines decreased while cell viability was preserved. Our findings indicate that even low-dose radiation can induce sterile inflammation by increasing serum HMGB1 and pro-inflammatory cytokines and that GL can ameliorate the sterile inflammatory process by inhibiting HMGB1 to preserve cell viability.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Glycyrrhizic Acid/pharmacology , Radiation Injuries, Experimental/drug therapy , Spleen/drug effects , Animals , Anti-Inflammatory Agents/therapeutic use , Cell Survival , Cells, Cultured , Cytokines/blood , Glycyrrhizic Acid/therapeutic use , HMGB1 Protein/antagonists & inhibitors , Male , Mice , Mice, Inbred BALB C , Oxidative Stress , Radiation Injuries, Experimental/prevention & control , Radiation, Ionizing , Spleen/radiation effects
17.
J Med Chem ; 64(18): 13439-13450, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34510899

ABSTRACT

During inflammatory reactions, the production and release of chemotactic factors guide the recruitment of selective leukocyte subpopulations. The alarmin HMGB1 and the chemokine CXCL12, both released in the microenvironment, can form a heterocomplex, which exclusively acts on the chemokine receptor CXCR4, enhancing cell migration, and in some pathological conditions such as rheumatoid arthritis exacerbates the immune response. An excessive cell influx at the inflammatory site can be diminished by disrupting the heterocomplex. Here, we report the computationally driven identification of the first peptide (HBP08) binding HMGB1 and selectively inhibiting the activity of the CXCL12/HMGB1 heterocomplex. Furthermore, HBP08 binds HMGB1 with the highest affinity reported so far (Kd of 0.8 ± 0.4 µM). The identification of this peptide represents an important step toward the development of innovative pharmacological tools for the treatment of severe chronic inflammatory conditions characterized by an uncontrolled immune response.


Subject(s)
Chemokine CXCL12/antagonists & inhibitors , HMGB1 Protein/antagonists & inhibitors , Peptides/pharmacology , Protein Binding/drug effects , Amino Acid Sequence , Animals , Cell Line , Cell Movement/drug effects , Chemokine CXCL12/metabolism , HMGB1 Protein/metabolism , Humans , Mice , Molecular Docking Simulation , Peptides/metabolism , Receptors, CXCR4/metabolism
18.
Neurotox Res ; 39(5): 1511-1523, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34417986

ABSTRACT

High mobility group box-1 (HMGB1) is a ubiquitous non-histone nuclear protein that plays a key role as a transcriptional activator, with its extracellular release provoking inflammation. Inflammatory responses are essential in methamphetamine (METH)-induced acute dopaminergic neurotoxicity. In the present study, we examined the effects of neutralizing anti-HMGB1 monoclonal antibody (mAb) on METH-induced dopaminergic neurotoxicity in mice. BALB/c mice received a single intravenous administration of anti-HMGB1 mAb prior to intraperitoneal injections of METH (4 mg/kg × 2, at 2-h intervals). METH injections induced hyperthermia, an increase in plasma HMGB1 concentration, degeneration of dopaminergic nerve terminals, accumulation of microglia, and extracellular release of neuronal HMGB1 in the striatum. These METH-induced changes were significantly inhibited by intravenous administration of anti-HMGB1 mAb. In contrast, blood-brain barrier disruption occurred by METH injections was not suppressed. Our findings demonstrated the neuroprotective effects of anti-HMGB1 mAb against METH-induced dopaminergic neurotoxicity, suggesting that HMGB1 could play an initially important role in METH toxicity.


Subject(s)
Antibodies, Monoclonal/pharmacology , Dopamine Uptake Inhibitors/toxicity , Dopaminergic Neurons/drug effects , HMGB1 Protein/antagonists & inhibitors , Methamphetamine/toxicity , Neuroprotective Agents/pharmacology , Animals , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , HMGB1 Protein/blood , Male , Mice , Mice, Inbred BALB C
19.
Biochem Biophys Res Commun ; 573: 164-170, 2021 10 08.
Article in English | MEDLINE | ID: mdl-34418763

ABSTRACT

During ischemia reperfusion (IR) injury, high mobility group box 1 (HMGB1), a chromatin binding protein, is released from necrotic cells and triggers inflammatory responses. We assessed the therapeutic effect of a neutralizing anti-HMGB1 monoclonal antibody (mAb) on lung IR injury. A murine hilar clamp model of IR was used, where mice were divided into sham and IR groups with intravenous administration of anti-HMGB 1 mAb or control mAb. We analyzed the effect of anti-HMGB1 mAb against IR injury by assessing lung oxygenation, lung injury score, neutrophil infiltration, expression of proinflammatory cytokines and chemokines, levels of mitogen-activated protein kinase (MAPK) signaling, and measurement of apoptotic cells. Anti-HMGB1 mAb significantly decreased the plasma level of HMGB1 elevated by IR. The severity of IR injury represented by oxygenation capacity, lung injury score, and neutrophil infiltration was significantly improved by anti-HMGB1 mAb treatment. The expression of proinflammatory factors, including IL-1ß, IL-6, IL-12, TNF-α, CXCL-1, and CXCL-2, and phosphorylation of p38 MAPK were both significantly reduced by anti-HMGB1 mAb treatment. Furthermore, anti-HMGB1 mAb treatment suppressed apoptosis, as determined through TUNEL assays. Overall, anti-HMGB1 mAb ameliorated lung IR injury by reducing inflammatory responses and apoptosis. Our findings indicate that anti-HMGB1 mAb has potential for use as a therapeutic to improve IR injury symptoms during lung transplantation.


Subject(s)
Antibodies, Monoclonal/metabolism , HMGB1 Protein/antagonists & inhibitors , Protective Agents/pharmacology , Reperfusion Injury/drug therapy , Animals , Apoptosis/drug effects , Female , HMGB1 Protein/metabolism , Mice , Mice, Inbred C57BL , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
20.
Inflamm Res ; 70(10-12): 1101-1111, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34455489

ABSTRACT

OBJECTIVE: High mobility group box-1 (HMGB1) has been reported to be involved in influenza A virus-induced acute respiratory distress syndrome (ARDS). We studied the efficacy of an anti-HMGB1 mAb using an in vitro model of TNF-α stimulation or influenza A virus infection in human pulmonary microvascular endothelial cells (HMVECs). METHODS: Vascular permeability of HMVECs was quantified using the Boyden chamber assay under tumor necrosis factor-α (TNF-α) stimulation or influenza A virus infection in the presence of anti-HMGB1 mAb or control mAb. The intracellular localization of HMGB1 was assessed by immunostaining. Extracellular cytokine concentrations and intracellular viral mRNA expression were quantified by the enzyme-linked immunosorbent assay and quantitative reverse transcription PCR, respectively. RESULTS: Vascular permeability was increased by TNF-α stimulation or influenza A infection; HMVECs became elongated and the intercellular gaps were extended. Anti-HMGB1 mAb suppressed both the increase in permeability and the cell morphology changes. Translocation of HMGB1 to the cytoplasm was observed in the non-infected cells. Although anti-HMGB1 mAb did not suppress viral replication, it did suppress cytokine production in HMVECs. CONCLUSION: Anti-HMGB1 mAb might be an effective therapy for severe influenza ARDS.


Subject(s)
Antibodies, Monoclonal/pharmacology , Capillary Permeability/drug effects , Cytokines/immunology , Endothelial Cells/drug effects , HMGB1 Protein/antagonists & inhibitors , Influenza A Virus, H3N2 Subtype , Influenza, Human/immunology , Animals , Cells, Cultured , Dogs , Endothelial Cells/metabolism , Endothelial Cells/virology , HMGB1 Protein/immunology , Humans , Lung/cytology
SELECTION OF CITATIONS
SEARCH DETAIL