Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.142
Filtrer
1.
Redox Biol ; 76: 103342, 2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-39265498

RÉSUMÉ

BACKGROUND: Disruption of the blood-brain barrier (BBB) is a major contributor to hemorrhagic transformation (HT) in patients with acute ischemic stroke (AIS) following intravenous thrombolysis (IVT). However, the clinical therapies aimed at BBB protection after IVT remain limited. METHODS: One hundred patients with AIS who underwent IVT were enrolled (42 with HT and 58 without HT 24 h after IVT). Based on the cytokine chip, the serum levels of several AIS-related proteins, including LCN2, ferritin, matrix metalloproteinase-3, vascular endothelial-derived growth factor, and X-linked inhibitor of apoptosis, were detected upon admission, and their associations with HT were analyzed. After finding that LCN2 was related to HT in patients with IVT, we clarified whether the modulation of LCN2 influenced BBB dysfunction and HT after thrombolysis and investigated the potential mechanism. RESULTS: In patients with AIS following IVT, logistic regression analysis showed that baseline serum LCN2 (p = 0.023) and ferritin (p = 0.046) levels were independently associated with HT. A positive correlation between serum LCN2 and ferritin levels was identified in patients with HT. In experimental studies, recombinant LCN2 (rLCN2) significantly aggravated BBB dysfunction and HT in the thromboembolic stroke rats after thrombolysis, whereas LCN2 inhibition by ZINC006440089 exerted opposite effects. Further mechanistic studies showed that, LCN2 promoted endothelial cell ferroptosis, accompanied by the induction of high mobility group box 1 (HMGB1) and the inhibition of nuclear translocation of nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) proteins. Ferroptosis inhibitor ferrostatin-1 (fer-1) significantly restricted the LCN2-mediated BBB disruption. Transfection of LCN2 and HMGB1 siRNA inhibited the endothelial cell ferroptosis, and this effects was reversed by Nrf2 siRNA. CONCLUSION: LCN2 aggravated BBB disruption after thrombolysis by promoting endothelial cell ferroptosis via regulating the HMGB1/Nrf2/HO-1 pathway, this may provide a promising therapeutic target for the prevention of HT after IVT.

2.
Brain Behav Immun ; 122: 583-595, 2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-39222725

RÉSUMÉ

Damage-associated molecular patterns (DAMPs) are endogenous molecules released in tissues upon cellular damage and necrosis, acting to initiate sterile inflammation. Constitutive DAMPs (cDAMPs) have the particularity to be present within the intracellular compartments of healthy cells, where they exert diverse functions such as regulation of gene expression and cellular homeostasis. However, after injury to the central nervous system (CNS), cDAMPs are rapidly released by stressed, damaged or dying neuronal, glial and endothelial cells, and can trigger inflammation without undergoing structural modifications. Several cDAMPs have been described in the injured CNS, such as interleukin (IL)-1α, IL-33, nucleotides (e.g. ATP), and high-mobility group box protein 1. Once in the extracellular milieu, these molecules are recognized by the remaining surviving cells through specific DAMP-sensing receptors, thereby inducing a cascade of molecular events leading to the production and release of proinflammatory cytokines and chemokines, as well as cell adhesion molecules. The ensuing immune response is necessary to eliminate cellular debris caused by the injury, allowing for damage containment. However, seeing as some molecules associated with the inflammatory response are toxic to surviving resident CNS cells, secondary damage occurs, aggravating injury and exacerbating neurological and behavioral deficits. Thus, a better understanding of these cDAMPs, as well as their receptors and downstream signaling pathways, could lead to identification of novel therapeutic targets for treating CNS injuries such as SCI, TBI, and stroke. In this review, we summarize the recent literature on cDAMPs, their specific functions, and the therapeutic potential of interfering with cDAMPs or their signaling pathways.


Sujet(s)
Alarmines , Système nerveux central , Humains , Alarmines/métabolisme , Animaux , Système nerveux central/métabolisme , Système nerveux central/traumatismes , Inflammation/métabolisme , Cytokines/métabolisme , Protéine HMGB1/métabolisme , Interleukine-33/métabolisme , Interleukine-1 alpha/métabolisme , Transduction du signal/physiologie
3.
Nanotoxicology ; : 1-17, 2024 Sep 19.
Article de Anglais | MEDLINE | ID: mdl-39295432

RÉSUMÉ

We have previously demonstrated that exposure to cobalt nanoparticles (Nano-Co) caused extensive interstitial fibrosis and inflammatory cell infiltration in mouse lungs. However, the underlying mechanisms of Nano-Co-induced pulmonary fibrosis remain unclear. In this study, we investigated the role of high-mobility group box 1 (HMGB1) in the epithelial cell-fibroblast crosstalk in Nano-Co-induced pulmonary fibrosis. Our results showed that Nano-Co exposure caused remarkable production and release of HMGB1, as well as nuclear accumulation of HIF-1α in human bronchial epithelial cells (BEAS-2B) in a dose- and a time-dependent manner. Pretreatment with CAY10585, an inhibitor against HIF-1α, significantly blocked the overexpression of HMGB1 in cell lysate and the release of HMGB1 in the supernatant of BEAS-2B cells induced by Nano-Co exposure, indicating that Nano-Co exposure induces HIF-1α-dependent HMGB1 overexpression and release. In addition, treatment of lung fibroblasts (MRC-5) with conditioned media from Nano-Co-exposed BEAS-2B cells caused increased RAGE expression, MAPK signaling activation, and enhanced expression of fibrosis-associated proteins, such as fibronectin, collagen 1, and α-SMA. However, conditioned media from Nano-Co-exposed BEAS-2B cells with HMGB1 knockdown had no effects on the activation of MRC-5 fibroblasts. Finally, inhibition of ERK1/2, p38, and JNK all abolished MRC-5 activation induced by conditioned media from Nano-Co-exposed BEAS-2B cells, suggesting that MAPK signaling might be a key downstream signal of HMGB1/RAGE to promote MRC-5 fibroblast activation. These findings have important implications for understanding the pro-fibrotic potential of Nano-Co.

4.
Sci Rep ; 14(1): 21421, 2024 09 13.
Article de Anglais | MEDLINE | ID: mdl-39271757

RÉSUMÉ

Traumatic brain injury (TBI) has been found to be associated with certain peripheral organ injuries; however, a few studies have explored the chronological influences of TBI on multiple organs and the systemic effects of therapeutic interventions. Particularly, high-mobility group box 1 (HMGB1) is a potential therapeutic target for TBI; however, its effects on peripheral organs remain unclear. Therefore, this study aimed to determine whether severe TBI can lead to multiple organ injury and how HMGB1 inhibition affects peripheral organs. This study used a weight drop-induced TBI mouse model and found that severe TBI can trigger short-lived systemic inflammation, in the lungs and liver, but not in the kidneys, regardless of the severity of the injury. TBI led to an increase in circulating HMGB1 and enhanced gene expressions of its receptors in every organ. Anti-HMGB1 antibody treatment reduced neuroinflammation but increased inflammation in peripheral organs. This study also found that HMGB1 inhibition appears to have a beneficial role in early neuroinflammation but could lead to detrimental effects on peripheral organs through decreased peripheral immune suppression. This study provides novel insights into the chronological changes in multiple organs due to TBI and the unique roles of HMGB1 between the brain and other organs.


Sujet(s)
Lésions traumatiques de l'encéphale , Modèles animaux de maladie humaine , Protéine HMGB1 , Protéine HMGB1/métabolisme , Animaux , Lésions traumatiques de l'encéphale/métabolisme , Lésions traumatiques de l'encéphale/anatomopathologie , Souris , Mâle , Souris de lignée C57BL , Rein/métabolisme , Rein/anatomopathologie , Foie/métabolisme , Foie/anatomopathologie , Foie/traumatismes , Inflammation/métabolisme , Poumon/anatomopathologie , Poumon/métabolisme
5.
World J Diabetes ; 15(9): 1979-2001, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-39280179

RÉSUMÉ

BACKGROUND: Diabetic intracerebral hemorrhage (ICH) is a serious complication of diabetes. The role and mechanism of bone marrow mesenchymal stem cell (BMSC)-derived exosomes (BMSC-exo) in neuroinflammation post-ICH in patients with diabetes are unknown. In this study, we investigated the regulation of BMSC-exo on hyperglycemia-induced neuroinflammation. AIM: To study the mechanism of BMSC-exo on nerve function damage after diabetes complicated with cerebral hemorrhage. METHODS: BMSC-exo were isolated from mouse BMSC media. This was followed by transfection with microRNA-129-5p (miR-129-5p). BMSC-exo or miR-129-5p-overexpressing BMSC-exo were intravitreally injected into a diabetes mouse model with ICH for in vivo analyses and were cocultured with high glucose-affected BV2 cells for in vitro analyses. The dual luciferase test and RNA immunoprecipitation test verified the targeted binding relationship between miR-129-5p and high-mobility group box 1 (HMGB1). Quantitative polymerase chain reaction, western blotting, and enzyme-linked immunosorbent assay were conducted to assess the levels of some inflammation factors, such as HMGB1, interleukin 6, interleukin 1ß, toll-like receptor 4, and tumor necrosis factor α. Brain water content, neural function deficit score, and Evans blue were used to measure the neural function of mice. RESULTS: Our findings indicated that BMSC-exo can promote neuroinflammation and functional recovery. MicroRNA chip analysis of BMSC-exo identified miR-129-5p as the specific microRNA with a protective role in neuroinflammation. Overexpression of miR-129-5p in BMSC-exo reduced the inflammatory response and neurological impairment in comorbid diabetes and ICH cases. Furthermore, we found that miR-129-5p had a targeted binding relationship with HMGB1 mRNA. CONCLUSION: We demonstrated that BMSC-exo can reduce the inflammatory response after ICH with diabetes, thereby improving the neurological function of the brain.

6.
Curr Issues Mol Biol ; 46(8): 8995-9015, 2024 Aug 17.
Article de Anglais | MEDLINE | ID: mdl-39194749

RÉSUMÉ

Atherosclerosis is a condition that is associated with lipid accumulation in the arterial intima. Consequently, the enlarging lesion, which is also known as an atherosclerotic plaque, may close the blood vessel lumen, thus leading to organ ischaemia. Furthermore, the plaque may rupture and initiate the formation of a thrombus, which can cause acute ischaemia. Atherosclerosis is a background pathological condition that can eventually lead to major cardiovascular diseases such as acute coronary syndrome or ischaemic stroke. The disorder is associated with an altered profile of alarmins, stress response molecules that are secreted due to cell injury or death and that induce inflammatory responses. High-mobility group box 1 (HMGB1), S100 proteins, interleukin-33, and heat shock proteins (HSPs) also affect the behaviour of endothelial cells and vascular smooth muscle cells (VSMCs). Thus, alarmins control the inflammatory responses of endothelial cells and proliferation of VSMCs, two important processes implicated in the pathogenesis of atherosclerosis. In this review, we will discuss the role of alarmins in the pathophysiology of atherosclerosis and myocardial infarction.

7.
Vet Immunol Immunopathol ; 275: 110815, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39153273

RÉSUMÉ

The objective of this study was to examine the values of MX dynamin-like GTPase 1 (Mx1), high mobility group box-1 (HMGB1), systemic inflammatory response index (SIRI), systemic inflammatory index (SII), tumor necrosis factor (TNF), and other hematological indices in calves with systemic inflammatory response syndrome (SIRS). The study material was divided into two groups: the SIRS group (comprising 13 calves) and the control group (comprising 10 calves). The independent samples t-test and Mann-Whitney U test were employed for normally distributed and non-normally distributed data, respectively. The relationship between the two groups was determined using Spearman correlation coefficient analysis. Significant differences were identified between the SIRS group and the control group with regard to white blood cell (WBC; P < 0.05), neutrophil (NEU; P < 0.01), and neutrophil-to-lymphocyte ratio (NLR; P < 0.001) values, in addition to SIRI (P < 0.05), SII (P < 0.01) values. Furthermore, HMGB1 (P < 0.001), Mx1 (P < 0.05), and TNF values (P < 0.001) demonstrated notable disparities between the two groups. As a result of this study, it was concluded that there were significant increases in inflammatory hematological indices, as well as in the levels of HMGB1, Mx1, and TNF, in calves with SIRS.


Sujet(s)
Animaux nouveau-nés , Maladies des bovins , Diarrhée , Protéine HMGB1 , Protéines de résistance aux myxovirus , Syndrome de réponse inflammatoire généralisée , Facteur de nécrose tumorale alpha , Animaux , Protéine HMGB1/sang , Bovins , Syndrome de réponse inflammatoire généralisée/médecine vétérinaire , Syndrome de réponse inflammatoire généralisée/sang , Syndrome de réponse inflammatoire généralisée/immunologie , Protéines de résistance aux myxovirus/génétique , Facteur de nécrose tumorale alpha/sang , Maladies des bovins/immunologie , Maladies des bovins/sang , Animaux nouveau-nés/immunologie , Diarrhée/médecine vétérinaire , Diarrhée/immunologie , Mâle , Femelle , Inflammation/médecine vétérinaire , Inflammation/sang , Inflammation/immunologie
8.
World J Gastrointest Surg ; 16(7): 2003-2011, 2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-39087106

RÉSUMÉ

BACKGROUND: Necrotising enterocolitis (NEC) is a critical gastrointestinal emergency affecting premature and low-birth-weight neonates. Serum amyloid A (SAA), procalcitonin (PCT), and high-mobility group box 1 (HMGB1) have emerged as potential biomarkers for NEC due to their roles in inflammatory response, tissue damage, and immune regulation. AIM: To evaluate the diagnostic value of SAA, PCT, and HMGB1 in the context of NEC in newborns. METHODS: The study retrospectively analysed the clinical data of 48 newborns diagnosed with NEC and 50 healthy newborns admitted to the hospital. Clinical, radiological, and laboratory findings, including serum SAA, PCT, and HMGB1 Levels, were collected, and specific detection methods were used. The diagnostic value of the biomarkers was evaluated through statistical analysis, which was performed using chi-square test, t-test, correlation analysis, and receiver operating characteristic (ROC) analysis. RESULTS: The study demonstrated significantly elevated levels of serum SAA, PCT, and HMGB1 Levels in newborns diagnosed with NEC compared with healthy controls. The correlation analysis indicated strong positive correlations among serum SAA, PCT, and HMGB1 Levels and the presence of NEC. ROC analysis revealed promising sensitivity and specificity for serum SAA, PCT, and HMGB1 Levels as potential diagnostic markers. The combined model of the three biomarkers demonstrating an extremely high area under the curve (0.908). CONCLUSION: The diagnostic value of serum SAA, PCT, and HMGB1 Levels in NEC was highlighted. These biomarkers potentially improve the early detection, risk stratification, and clinical management of critical conditions. The findings suggest that these biomarkers may aid in timely intervention and the enhancement of outcomes for neonates affected by NEC.

9.
Front Cell Dev Biol ; 12: 1348707, 2024.
Article de Anglais | MEDLINE | ID: mdl-39100092

RÉSUMÉ

Background: Cancer cell evasion of the immune response is critical to cancer development and metastases. Clinicians' ability to kickstart the immune system to target these rogue cells is an ever-growing area of research and medicine. This study delved into the relationship between lipid metabolism, High Mobility Group Box 1 protein (HMGB1)-a pro-inflammatory damage-associated molecular pattern protein-and immune regulation within non-small cell lung adenocarcinoma (NSCLC). Method: To address this question, we used a combination of proteomics, molecular biology, and bioinformatic techniques to investigate the relationship between fatty acids and immune signals within NSCLC. Results: We found that the expression of stearoyl CoA desaturase 1 (SCD1) was decreased in NSCLC tumors compared to normal tissues. This emphasized the critical role of lipid metabolism in tumor progression. Interestingly, monounsaturated fatty acid (MUFA) availability affected the expression of programmed death ligand-1 (PD-L1), a pivotal immune checkpoint target in lung cancer cells and immune cells, as well as HMGB1, suggesting a novel approach to modulating the immune response. This study uncovered a complex interplay between SCD1, PD-L1, and HMGB1, influencing the immunological sensitivity of tumors. Conclusion: Our work underscores the critical importance of understanding the intricate relationships between lipid metabolism and immune modulation to develop more effective NSCLC treatments and personalized therapies. As we continue to explore these connections, we hope to contribute significantly to the ever-evolving field of cancer research, improving patient outcomes and advancing precision medicine in NSCLC.

10.
Regen Ther ; 26: 415-424, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-39070123

RÉSUMÉ

Introduction: This present study evaluated the effect of combination therapy with stromal cell-derived factor 1α (SDF-1α) and high-mobility group box 1 (HMGB1) peptide on the regeneration of tracheal injury in a rat model. Methods: To improve this effect, SDF-1α was incorporated into a gelatin hydrogel, which was then applied to the damaged tracheal cartilage of rats for local release. Furthermore, HMGB1 peptide was repeatedly administered intravenously. Regeneration of damaged tracheal cartilage was evaluated in terms of cell recruitment. Results: Mesenchymal stem cells (MSC) with C-X-C motif chemokine receptor 4 (CXCR4) were mobilized more into the injured area, and consequently the fastest tracheal cartilage regeneration was observed in the combination therapy group eight weeks after injury. Conclusions: The present study demonstrated that combination therapy with gelatin hydrogel incorporating SDF-1α and HMGB1 peptide injected intravenously can enhance the recruitment of CXCR4-positive MSC, promoting the regeneration of damaged tracheal cartilage.

11.
Int J Mol Sci ; 25(11)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38892032

RÉSUMÉ

Keloids, marked by abnormal cellular proliferation and excessive extracellular matrix (ECM) accumulation, pose significant therapeutic challenges. Ethyl pyruvate (EP), an inhibitor of the high-mobility group box 1 (HMGB1) and TGF-ß1 pathways, has emerged as a potential anti-fibrotic agent. Our research evaluated EP's effects on keloid fibroblast (KF) proliferation and ECM production, employing both in vitro cell cultures and ex vivo patient-derived keloid spheroids. We also analyzed the expression levels of ECM components in keloid tissue spheroids treated with EP through immunohistochemistry. Findings revealed that EP treatment impedes the nuclear translocation of HMGB1 and diminishes KF proliferation. Additionally, EP significantly lowered mRNA and protein levels of collagen I and III by attenuating TGF-ß1 and pSmad2/3 complex expression in both human dermal fibroblasts and KFs. Moreover, metalloproteinase I (MMP-1) and MMP-3 mRNA levels saw a notable increase following EP administration. In keloid spheroids, EP induced a dose-dependent reduction in ECM component expression. Immunohistochemical and western blot analyses confirmed significant declines in collagen I, collagen III, fibronectin, elastin, TGF-ß, AKT, and ERK 1/2 expression levels. These outcomes underscore EP's antifibrotic potential, suggesting its viability as a therapeutic approach for keloids.


Sujet(s)
Fibroblastes , Chéloïde , Pyruvates , Sphéroïdes de cellules , Humains , Chéloïde/métabolisme , Chéloïde/anatomopathologie , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Pyruvates/pharmacologie , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Sphéroïdes de cellules/métabolisme , Matrix metalloproteinase 1/métabolisme , Matrix metalloproteinase 1/génétique , Facteur de croissance transformant bêta-1/métabolisme , Protéine HMGB1/métabolisme , Protéine HMGB1/génétique , Collagène/métabolisme , Collagène/biosynthèse , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Matrix metalloproteinase 3/métabolisme , Matrix metalloproteinase 3/génétique , Matrice extracellulaire/métabolisme , Matrice extracellulaire/effets des médicaments et des substances chimiques , Collagène de type I/métabolisme , Collagène de type I/génétique , Protéine Smad2/métabolisme , Protéine Smad2/génétique , Protéine Smad-3/métabolisme , Régulation positive/effets des médicaments et des substances chimiques , Mâle
12.
Food Chem Toxicol ; 190: 114762, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38871110

RÉSUMÉ

In recent years, carbonized silicon nanoparticles (SiC NPs) have found widespread scientific and engineering applications, raising concerns about potential human health risks. SiC NPs may induce pulmonary damage through sustained inflammatory responses and oxidative stress, with unclear toxicity mechanisms. This study uses an in vitro co-culture model of alveolar macrophages (NR8383) and alveolar epithelial cells (RLE-6TN) to simulate the interaction between airway epithelial cells and immune cells, providing initial insights into SiC NP-triggered inflammatory responses. The research reveals that increasing SiC NP exposure prompts NR8383 cells to release high mobility group box 1 protein (HMGB1), which migrates into RLE-6TN cells and activates the receptor for advanced glycation end-products (RAGE) and Toll-like receptor 4 (TLR4). RAGE and TLR4 synergistically activate the MyD88/NF-κB inflammatory pathway, ultimately inducing inflammatory responses and oxidative stress in RLE-6TN cells, characterized by excessive ROS generation and altered cytokine levels. Pretreatment with RAGE and TLR4 inhibitors attenuates SiC-induced HMGB1 expression and downstream pathway proteins, reducing inflammatory responses and oxidative damage. This highlights the pivotal role of RAGE-TLR4 crosstalk in SiC NP-induced pulmonary inflammation, providing insights into SiC NP cytotoxicity and nanomaterial safety guidelines.


Sujet(s)
Cellules épithéliales , Protéine HMGB1 , Poumon , Macrophages alvéolaires , Nanoparticules , Composés du silicium , Animaux , Humains , Rats , Composés inorganiques du carbone/toxicité , Lignée cellulaire , Techniques de coculture , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Protéine HMGB1/métabolisme , Inflammation/induit chimiquement , Inflammation/métabolisme , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Macrophages alvéolaires/effets des médicaments et des substances chimiques , Macrophages alvéolaires/métabolisme , Nanoparticules/toxicité , Stress oxydatif/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Récepteur spécifique des produits finaux de glycosylation avancée/métabolisme , Composés du silicium/toxicité , Récepteur de type Toll-4/métabolisme
13.
Cell Mol Gastroenterol Hepatol ; 18(3): 101362, 2024.
Article de Anglais | MEDLINE | ID: mdl-38788899

RÉSUMÉ

BACKGROUND & AIMS: There is limited information on how the liver-to-gut axis contributes to alcohol-associated liver disease (AALD). We previously identified that high-mobility group box-1 (HMGB1) undergoes oxidation in hepatocytes and demonstrated elevated serum levels of oxidized HMGB1 ([O] HMGB1) in alcoholic patients. Since interleukin-1 beta (IL-1B) increases in AALD, we hypothesized hepatocyte-derived [O] HMGB1 could interact with IL-1B to activate a pro-inflammatory program that, besides being detrimental to the liver, drives intestinal barrier dysfunction. RESULTS: Alcohol-fed RageΔMye mice exhibited decreased nuclear factor kappa B signaling, a pro-inflammatory signature, and reduced total intestinal permeability, resulting in protection from AALD. In addition, [O] HMGB1 bound and signaled through the receptor for advanced-glycation end-products (RAGE) in myeloid cells, driving hepatic inflammation, intestinal permeability, and increased portal blood lipopolysaccharide in AALD. We identified that [O] HMGB1 formed a complex with IL-1B, which was found in the livers of patients with acute alcoholic hepatitis and mice with AALD. This complex originated from the liver, because it was absent in the intestine when hepatocytes did not produce [O] HMGB1. Mechanistically, the complex bound RAGE in Kupffer cells and macrophages induced a pro-inflammatory program. Moreover, it bound RAGE in intestinal macrophages and epithelial cells, leading to intestinal inflammation, altered intestinal epithelial cell tight junction protein expression, increased intestinal permeability, and elevated portal blood lipopolysaccharide, enhancing AALD pathogenesis. CONCLUSIONS: We identified a protein complex of liver origin that amplifies the pro-inflammatory feedback loop in AALD; therefore, targeting this complex could have significant therapeutic potential.


Sujet(s)
Protéine HMGB1 , Interleukine-1 bêta , Maladies alcooliques du foie , Foie , Récepteur spécifique des produits finaux de glycosylation avancée , Animaux , Protéine HMGB1/métabolisme , Souris , Maladies alcooliques du foie/anatomopathologie , Maladies alcooliques du foie/métabolisme , Humains , Récepteur spécifique des produits finaux de glycosylation avancée/métabolisme , Foie/métabolisme , Foie/anatomopathologie , Interleukine-1 bêta/métabolisme , Mâle , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Modèles animaux de maladie humaine , Transduction du signal , Intestins/anatomopathologie , Perméabilité , Facteur de transcription NF-kappa B/métabolisme , Souris de lignée C57BL , Inflammation/métabolisme , Inflammation/anatomopathologie
14.
Biochim Biophys Acta Rev Cancer ; 1879(3): 189105, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38701938

RÉSUMÉ

The present study explores the complex roles of High Mobility Group Box 1 (HMGB1) in the context of cancer development, emphasizing glioblastoma (GBM) and other central nervous system (CNS) cancers. HMGB1, primarily known for its involvement in inflammation and angiogenesis, emerges as a multifaceted player in the tumorigenesis of GBM. The overexpression of HMGB1 correlates with glioma malignancy, influencing key pathways like RAGE/MEK/ERK and RAGE/Rac1. Additionally, HMGB1 secretion is linked to the maintenance of glioma stem cells (GSCs) and contributes to the tumor microenvironment's (TME) vascular leakiness. Henceforth, our review discusses the bidirectional impact of HMGB1, acting as both a promoter of tumor progression and a mediator of anti-tumor immune responses. Notably, HMGB1 exhibits tumor-suppressive roles by inducing apoptosis, limiting cellular proliferation, and enhancing the sensitivity of GBM to therapeutic interventions. This dualistic nature of HMGB1 calls for a nuanced understanding of its implications in GBM pathogenesis, offering potential avenues for more effective and personalized treatment strategies. The findings underscore the need to explore HMGB1 as a prognostic marker, therapeutic target, and a promising tool for stimulating anti-tumor immunity in GBM.


Sujet(s)
Tumeurs du système nerveux central , Glioblastome , Protéine HMGB1 , Microenvironnement tumoral , Humains , Protéine HMGB1/métabolisme , Glioblastome/anatomopathologie , Glioblastome/traitement médicamenteux , Glioblastome/métabolisme , Glioblastome/immunologie , Tumeurs du système nerveux central/métabolisme , Tumeurs du système nerveux central/anatomopathologie , Tumeurs du système nerveux central/traitement médicamenteux , Animaux , Cellules souches tumorales/anatomopathologie , Cellules souches tumorales/métabolisme , Transduction du signal , Prolifération cellulaire
15.
Int J Mol Sci ; 25(10)2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38791199

RÉSUMÉ

Danger-associated molecular patterns (DAMPs) are elevated within the amniotic cavity, and their increases correlate with advancing gestational age, chorioamnionitis, and labor. Although the specific triggers for their release in utero remain unclear, it is thought that they may contribute to the initiation of parturition by influencing cellular stress mechanisms that make the fetal membranes (FMs) more susceptible to rupture. DAMPs induce inflammation in many different tissue types. Indeed, they precipitate the subsequent release of several proinflammatory cytokines that are known to be key for the weakening of FMs. Previously, we have shown that in vitro stretch of human amnion epithelial cells (hAECs) induces a cellular stress response that increases high-mobility group box-1 (HMGB1) secretion. We have also shown that cell-free fetal DNA (cffDNA) induces a cytokine response in FM explants that is fetal sex-specific. Therefore, the aim of this work was to further investigate the link between stretch and the DAMPs HMGB1 and cffDNA in the FM. These data show that stretch increases the level of cffDNA released from hAECs. It also confirms the importance of the sex of the fetus by demonstrating that female cffDNA induced more cellular stress than male fetuses. Our data treating hAECs and human amnion mesenchymal cells with HMGB1 show that it has a differential effect on the ability of the cells of the amnion to upregulate the proinflammatory cytokines and propagate a proinflammatory signal through the FM that may weaken it. Finally, our data show that sulforaphane (SFN), a potent activator of Nrf2, is able to mitigate the proinflammatory effects of stretch by decreasing the levels of HMGB1 release and ROS generation after stretch and modulating the increase of key cytokines after cell stress. HMGB1 and cffDNA are two of the few DAMPs that are known to induce cytokine release and matrix metalloproteinase (MMP) activation in the FMs; thus, these data support the general thesis that they can function as potential central players in the normal mechanisms of FM weakening during the normal distension of this tissue at the end of a normal pregnancy.


Sujet(s)
Membranes extraembryonnaires , Protéine HMGB1 , Inflammation , Humains , Protéine HMGB1/métabolisme , Protéine HMGB1/génétique , Femelle , Grossesse , Inflammation/métabolisme , Inflammation/anatomopathologie , Membranes extraembryonnaires/métabolisme , Acides nucléiques acellulaires/métabolisme , Mâle , Amnios/métabolisme , Cytokines/métabolisme , Cellules épithéliales/métabolisme , Cellules cultivées , Alarmines/métabolisme
16.
J Gastroenterol ; 59(8): 744-757, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38727823

RÉSUMÉ

BACKGROUND: Chronic pancreatitis (CP) is a progressive disease characterized by pancreatic fibrosis for which effective treatment options are lacking. Mesenchymal stem cells (MSCs) have shown potential for fibrosis treatment but face limitations in clinical application. The high-mobility group box 1 (HMGB1) fragment mobilizes MSCs from bone marrow into the blood and has emerged as a promising therapeutic agent for tissue regeneration in various pathological conditions. The aim of this study was to investigate the potential therapeutic effects of systemic administration of the HMGB1 fragment in a mouse model of CP. METHODS: A caerulein-induced CP mouse model was used, and the HMGB1 fragment was administered by tail vein injection. Parameters such as body weight, pancreatic tissue damage, fibrosis, inflammatory cytokine expression, and collagen-related gene expression were evaluated using various assays, including immunohistochemistry, real-time PCR, serum analysis, and single-cell transcriptome analysis. And the migration of MSCs to the pancreas was evaluated using the parabiosis model. RESULTS: Administration of the HMGB1 fragment was associated with significant improvements in pancreatic tissue damage and fibrosis. It suppressed the expression of inflammatory cytokines and activated platelet-derived growth factor receptor-α+ MSCs, leading to their accumulation in the pancreas. The HMGB1 fragment also shifted gene expression patterns associated with pancreatic fibrosis toward those of the normal pancreas. Systemic administration of the HMGB1 fragment demonstrated therapeutic efficacy in attenuating pancreatic tissue damage and fibrosis in a CP mouse model. CONCLUSION: These findings highlight the potential of the HMGB1 fragment as a therapeutic target for the treatment of CP.


Sujet(s)
Céruléine , Modèles animaux de maladie humaine , Fibrose , Protéine HMGB1 , Pancréas , Pancréatite chronique , Animaux , Pancréatite chronique/traitement médicamenteux , Protéine HMGB1/métabolisme , Protéine HMGB1/génétique , Souris , Mâle , Pancréas/anatomopathologie , Souris de lignée C57BL , Cytokines/métabolisme , Cellules souches mésenchymateuses/métabolisme , Transplantation de cellules souches mésenchymateuses/méthodes
17.
Exp Ther Med ; 27(6): 266, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38756909

RÉSUMÉ

Gestational hypertension (GH) is a common disorder during pregnancy that can cause adverse pregnancy outcomes. In the present study, magnesium sulfate (MgSO4) combined with labetalol was used for clinical treatment. Randomized controlled trial was conducted in 100 patients with GH, documented in the Department of Obstetrics and Gynecology (Taicang TCM Hospital) grouped into the experimental (Expt) and control (Ctrl) groups (n=50 cases/group). The Ctrl group was treated with MgSO4, whereas the Expt group was treated with MgSO4 + labetalol. The systolic blood pressure (SBP) and diastolic blood pressure (DBP) in the Expt group were not significantly different from those in the Ctrl group (P>0.05). By contrast, the SBP and DBP were significantly lower after treatment than those before treatment in both groups (P<0.05). Whole blood viscosity, plasma viscosity and hematocrit were significantly lower in the Expt group compared with those in the Ctrl group after treatment (P<0.05). High mobility group box-1 protein, homocysteine and serum cystatin C levels in the Expt group were also markedly lower than those in the Ctrl group after treatment (P<0.05). In the Expt group, the rate of spontaneous vaginal delivery was much higher, whereas the rates of cesarean section and postpartum hemorrhage were markedly lower than those in the Ctrl group (P<0.05). The occurrence of fetal intrauterine distress, placental abruption, neonatal asphyxia, premature birth and neonatal death were also significantly lower in the Expt group than those in the Ctrl group (P<0.05). In conclusion, MgSO4 + labetalol could improve inflammatory stress and the hemodynamics of patients with GH, and may have a marked antihypertensive effect. Thus, it may improve pregnancy outcome and reduce perinatal complications.

18.
Biochim Biophys Acta Gen Subj ; 1868(6): 130614, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38598971

RÉSUMÉ

BACKGROUND: Deregulation of cell death is a common characteristic of cancer, and resistance to this process often occurs in lung cancer. Understanding the molecular mechanisms underlying an aberrant cell death is important. Recent studies have emphasized the involvement of calmodulin-regulated spectrin-associated protein 3 (CAMSAP3) in lung cancer aggressiveness, its influence on cell death regulation remains largely unexplored. METHODS: CAMSAP3 was knockout in lung cancer cells using CRISPR-Cas9 system. Cell death and autophagy were evaluated using MTT and autophagic detection assays. Protein interactions were performed by proteomic analysis and immunoprecipitation. Protein expressions and their cytoplasmic localization were analyzed through immunoblotting and immunofluorescence techniques. RESULTS: This study reveals a significant correlation between low CAMSAP3 expression and poor overall survival rates in lung cancer patients. Proteomic analysis identified high mobility group box 1 (HMGB1) as a candidate interacting protein involved in the regulation of cell death. Treatment with trichostatin A (TSA), an inhibitor of histone deacetylases (HDACs) resulted in increased HMGB1 acetylation and its translocation to the cytoplasm and secretion, thereby inducing autophagic cell death. However, this process was diminished in CAMSAP3 knockout lung cancer cells. Mechanistically, immunoprecipitation indicated an interaction between CAMSAP3 and HMGB1, particularly with its acetylated form, in which this complex was elevated in the presence of TSA. CONCLUSIONS: CAMSAP3 is prerequisite for TSA-mediated autophagic cell death by interacting with cytoplasmic acetylated HMGB1 and enhancing its release. SIGNIFICANT: This finding provides molecular insights into the role of CAMSAP3 in regulating cell death, highlighting its potential as a therapeutic target for lung cancer treatment.


Sujet(s)
Protéine HMGB1 , Tumeurs du poumon , Humains , Protéine HMGB1/métabolisme , Protéine HMGB1/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Acétylation , Autophagie , Lignée cellulaire tumorale , Mort cellulaire , Cellules A549 , Acides hydroxamiques/pharmacologie
19.
Int Immunopharmacol ; 133: 112039, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38613884

RÉSUMÉ

BACKGROUND: Serum high mobility group box 1 (HMGB1) serves as a diagnostic biomarker for malignant peritoneal mesothelioma (MPM) patients, yet its diagnostic significance within MPM tumor tissues remains uncertain. This study aims to elucidate the roles of HMGB1 in MPM. METHODS: HMGB1 expression analysis was conducted in both tumor and adjacent non-cancerous tissues collected from MPM patients. The two-year follow-up of MPM patients commenced from the diagnosis date. Inflammatory cytokine analysis was performed on these tissues, and Pearson correlation coefficient analysis was applied to examine variable relationships. In vitro assays included constructing an HMGB1 knockdown cell line, assessing cell viability, apoptosis, and inflammatory cytokine levels to delineate HMGB1's roles in MPM. RESULTS: HMGB1 overexpression was observed in MPM tumor tissues, particularly in stages III-IV. Diagnostic implications of HMGB1 for MPM were evident, augmenting its diagnostic value. HMGB1 overexpression correlated with diminished survival rates. Positive correlations existed between inflammatory cytokines and HMGB1 in MPM tumor tissues and cell lines. Suppression of HMGB1 regulated cell growth and apoptosis in MPM cell lines. CONCLUSION: HMGB1 exhibits diagnostic potential for MPM and modulates inflammatory responses within the disease context.


Sujet(s)
Protéine HMGB1 , Mésothéliome malin , Tumeurs du péritoine , Adulte , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Apoptose , Marqueurs biologiques tumoraux/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire , Cytokines/métabolisme , Régulation de l'expression des gènes tumoraux , Protéine HMGB1/métabolisme , Protéine HMGB1/génétique , Inflammation/métabolisme , Tumeurs du péritoine/métabolisme
20.
Curr Issues Mol Biol ; 46(4): 3640-3675, 2024 Apr 19.
Article de Anglais | MEDLINE | ID: mdl-38666958

RÉSUMÉ

Alarmins are immune-activating factors released after cellular injury or death. By secreting alarmins, cells can interact with immune cells and induce a variety of inflammatory responses. The broad family of alarmins involves several members, such as high-mobility group box 1, S100 proteins, interleukin-33, and heat shock proteins, among others. Studies have found that the concentrations and expression profiles of alarmins are altered in immune-mediated diseases. Furthermore, they are involved in the pathogenesis of inflammatory conditions. The aim of this narrative review is to present the current evidence on the role of alarmins in rheumatoid arthritis, osteoarthritis, and psoriasis. We discuss their potential involvement in mechanisms underlying the progression of these diseases and whether they could become therapeutic targets. Moreover, we summarize the impact of pharmacological agents used in the treatment of these diseases on the expression of alarmins.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE