Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 669
Filter
1.
Georgian Med News ; (350): 120-126, 2024 May.
Article in English | MEDLINE | ID: mdl-39089283

ABSTRACT

The relationship between Helicobacter pylori infection and gallbladder diseases, particularly cholecystitis and gallbladder polyps, remains unclear. This study aimed to investigate the presence of H. pylori in gallbladder tissues and its potential role in gallbladder pathologies, as well as to examine the expression of chemokines CXCL2 and CXCL5 in these conditions. MATERIAL AND METHODS: A total of 137 laparoscopically excised gallbladders were analysed through histological examination, PCR for H. pylori-specific DNA, and quantitative real-time PCR for CXCL2 and CXCL5 gene expression. The study cohort included patients with acute calculous cholecystitis, chronic calculous cholecystitis, and gallbladder polyps. RESULTS: H. pylori was detected in 30.7% of cases by histological methods and 42.3% by PCR. Elevated expression of CXCL2 and CXCL5 was observed in 62% and 57.7% of cases, respectively, with a higher prevalence in acute cholecystitis compared to chronic conditions. However, no statistically significant association was found between H. pylori presence and the forms of cholecystitis, as well as between H. pylori presence and chemokine expression in gallbladder. CONCLUSIONS: The study did not establish a direct link between the presence of H. pylori infection and forms of gallbladder pathologies. The findings suggest that other factors other than H. pylori may contribute to the upregulation of CXCL2 and CXCL5 in gallbladder diseases. Further research is needed to elucidate the complex interactions between H. pylori, chemokines, and gallbladder pathologies.


Subject(s)
Chemokine CXCL2 , Chemokine CXCL5 , Gallbladder , Helicobacter Infections , Helicobacter pylori , Humans , Helicobacter pylori/isolation & purification , Helicobacter pylori/pathogenicity , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter Infections/complications , Helicobacter Infections/genetics , Male , Gallbladder/microbiology , Gallbladder/pathology , Gallbladder/surgery , Female , Middle Aged , Chemokine CXCL5/genetics , Chemokine CXCL5/metabolism , Chemokine CXCL2/genetics , Chemokine CXCL2/metabolism , Adult , Cholecystitis/microbiology , Cholecystitis/pathology , Cholecystitis/surgery , Polyps/microbiology , Polyps/pathology , Gallbladder Diseases/microbiology , Gallbladder Diseases/pathology , Gallbladder Diseases/surgery , Aged
2.
Biochem Pharmacol ; 226: 116379, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38908531

ABSTRACT

Sepsis is a widespread and life-threatening disease characterised by infection-triggered immune hyperactivation and cytokine storms, culminating in tissue damage and multiple organ dysfunction syndrome. BMAL1 is a pivotal transcription factor in the circadian clock that plays a crucial role in maintaining immune homeostasis. BMAL1 dysregulation has been implicated in inflammatory diseases and immunodeficiency. However, the mechanisms underlying BMAL1 disruption in sepsis-induced acute lung injury (ALI) remain poorly understood. In vitro, we used THP1 and mouse peritoneal macrophages to elucidate the potential mechanism of BMAL1 function in sepsis. In vivo, an endotoxemia model was used to investigate the effect of BMAL1 on sepsis and the therapeutic role of targeting CXCR2. We showed that BMAL1 significantly affected the regulation of innate immunity in sepsis-induced ALI. BMAL1 deficiency in the macrophages exacerbated systemic inflammation and sepsis-induced ALI. Mechanistically, BMAL1 acted as a transcriptional suppressor and regulated the expression of CXCL2. BMAL1 deficiency in macrophages upregulated CXCL2 expression, increasing the recruitment of polymorphonuclear neutrophils and the formation of neutrophil extracellular traps (NETs) by binding to the chemokine receptor CXCR2, thereby intensifying lung injury in a sepsis model. Furthermore, a selective inhibitor of CXCR2, SB225002, exerted promising therapeutic effects by markedly reducing neutrophil infiltration and NETs formation and alleviating lung injury. Importantly, CXCR2 blockade mitigated multiple organ dysfunction. Collectively, these findings suggest that BMAL1 controls the CXCL2/CXCR2 pathway, and the therapeutic efficacy of targeting CXCR2 in sepsis has been validated, presenting BMAL1 as a potential therapeutic target for lethal infections.


Subject(s)
ARNTL Transcription Factors , Acute Lung Injury , Homeostasis , Mice, Inbred C57BL , Receptors, Interleukin-8B , Sepsis , Animals , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Sepsis/immunology , Sepsis/metabolism , Sepsis/complications , Acute Lung Injury/etiology , Acute Lung Injury/immunology , Acute Lung Injury/metabolism , Mice , Humans , Receptors, Interleukin-8B/antagonists & inhibitors , Receptors, Interleukin-8B/metabolism , Receptors, Interleukin-8B/genetics , Homeostasis/physiology , Male , Mice, Knockout , Chemokine CXCL2/metabolism , THP-1 Cells
3.
PLoS Genet ; 20(6): e1011309, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38885192

ABSTRACT

PLK1 (Polo-like kinase 1) plays a critical role in the progression of lung adenocarcinoma (LUAD). Recent studies have unveiled that targeting PLK1 improves the efficacy of immunotherapy, highlighting its important role in the regulation of tumor immunity. Nevertheless, our understanding of the intricate interplay between PLK1 and the tumor microenvironment (TME) remains incomplete. Here, using genetically engineered mouse model and single-cell RNA-seq analysis, we report that PLK1 promotes an immunosuppressive TME in LUAD, characterized with enhanced M2 polarization of tumor associated macrophages (TAM) and dampened antigen presentation process. Mechanistically, elevated PLK1 coincides with increased secretion of CXCL2 cytokine, which promotes M2 polarization of TAM and diminishes expression of class II major histocompatibility complex (MHC-II) in professional antigen-presenting cells. Furthermore, PLK1 negatively regulates MHC-II expression in cancer cells, which has been shown to be associated with compromised tumor immunity and unfavorable patient outcomes. Taken together, our results reveal PLK1 as a novel modulator of TME in LUAD and provide possible therapeutic interventions.


Subject(s)
Adenocarcinoma of Lung , Cell Cycle Proteins , Lung Neoplasms , Polo-Like Kinase 1 , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins , Single-Cell Analysis , Tumor Microenvironment , Animals , Humans , Mice , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/pathology , Antigen Presentation/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chemokine CXCL2/genetics , Chemokine CXCL2/metabolism , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism
4.
Sci Adv ; 10(20): eadl6343, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38758783

ABSTRACT

Trauma rapidly mobilizes the immune response of surrounding tissues and activates regeneration program. Manipulating immune response to promote tissue regeneration shows a broad application prospect. However, the understanding of bone healing dynamics at cellular level remains limited. Here, we characterize the landscape of immune cells after alveolar bone injury and reveal a pivotal role of infiltrating natural killer T (NKT) cells. We observe a rapid increase in NKT cells after injury, which inhibit osteogenic differentiation of mesenchymal stem cells (MSCs) and impair alveolar bone healing. Cxcl2 is up-regulated in NKT cells after injury. Systemic administration of CXCL2-neutralizing antibody or genetic deletion of Cxcl2 improves the bone healing process. In addition, we fabricate a gelatin-based porous hydrogel to deliver NK1.1 depletion antibody, which successfully promotes alveolar bone healing. In summary, our study highlights the importance of NKT cells in the early stage of bone healing and provides a potential therapeutic strategy for accelerating bone regeneration.


Subject(s)
Bone Regeneration , Chemokine CXCL2 , Natural Killer T-Cells , Osteogenesis , Bone Regeneration/drug effects , Animals , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Mice , Osteogenesis/drug effects , Chemokine CXCL2/metabolism , Chemokine CXCL2/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Cell Differentiation , Mice, Inbred C57BL
5.
Int J Mol Sci ; 25(8)2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38674048

ABSTRACT

Inflammation processes of the central nervous system (CNS) play a vital role in the pathogenesis of several neurological and psychiatric disorders like depression. These processes are characterized by the activation of glia cells, such as microglia. Clinical studies showed a decrease in symptoms associated with the mentioned diseases after the treatment with anti-inflammatory drugs. Therefore, the investigation of novel anti-inflammatory drugs could hold substantial potential in the treatment of disorders with a neuroinflammatory background. In this in vitro study, we report the anti-inflammatory effects of a novel hexacyclic peptide-peptoid hybrid in lipopolysaccharide (LPS)-stimulated BV2 microglial cells. The macrocyclic compound X15856 significantly suppressed Interleukin 6 (IL-6), tumor necrosis factor-α (TNF-α), c-c motif chemokine ligand 2 (CCL2), CCL3, C-X-C motif chemokine ligand 2 (CXCL2), and CXCL10 expression and release in LPS-treated BV2 microglial cells. The anti-inflammatory effects of the compound are partially explained by the modulation of the phosphorylation of p38 mitogen-activated protein kinases (MAPK), p42/44 MAPK (ERK 1/2), protein kinase C (PKC), and the nuclear factor (NF)-κB, respectively. Due to its remarkable anti-inflammatory properties, this compound emerges as an encouraging option for additional research and potential utilization in disorders influenced by inflammation, such as depression.


Subject(s)
Anti-Inflammatory Agents , Lipopolysaccharides , Microglia , Microglia/drug effects , Microglia/metabolism , Animals , Mice , Anti-Inflammatory Agents/pharmacology , Cell Line , Peptoids/pharmacology , Peptoids/chemistry , Interleukin-6/metabolism , NF-kappa B/metabolism , Chemokine CCL2/metabolism , Chemokine CCL2/genetics , Peptides/pharmacology , Peptides/chemistry , Tumor Necrosis Factor-alpha/metabolism , Chemokine CXCL2/metabolism , Cytokines/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Chemokine CCL3/metabolism , Chemokine CCL3/genetics , Macrocyclic Compounds/pharmacology , Macrocyclic Compounds/chemistry
6.
Front Immunol ; 15: 1367019, 2024.
Article in English | MEDLINE | ID: mdl-38686389

ABSTRACT

Background: Although hyperuricemia is not always associated with acute gouty arthritis, uric acid is a significant risk factor for gout. Therefore, we investigated the specific mechanism of uric acid activity. Methods: Using the gout-associated transcriptome dataset GSE160170, we conducted differential expression analysis to identify differentially expressed genes (DEGs). Moreover, we discovered highly linked gene modules using weighted gene coexpression network analysis (WGCNA) and evaluated their intersection. Subsequently, we screened for relevant biomarkers using the cytoHubba and Mcode algorithms in the STRING database, investigated their connection to immune cells and constructed a competitive endogenous RNA (ceRNA) network to identify upstream miRNAs and lncRNAs. We also collected PBMCs from acute gouty arthritis patients and healthy individuals and constructed a THP-1 cell gout inflammatory model, RT-qPCR and western blotting (WB) were used to detect the expression of C-X-C motif ligand 8 (CXCL8), C-X-C motif ligand 2 (CXCL2), and C-X-C motif ligand 1 (CXCL1). Finally, we predicted relevant drug targets through hub genes, hoping to find better treatments. Results: According to differential expression analysis, there were 76 upregulated and 28 downregulated mRNAs in GSE160170. Additionally, WGCNA showed that the turquoise module was most strongly correlated with primary gout; 86 hub genes were eventually obtained upon intersection. IL1ß, IL6, CXCL8, CXCL1, and CXCL2 are the principal hub genes of the protein-protein interaction (PPI) network. Using RT-qPCR and WB, we found that there were significant differences in the expression levels of CXCL8, CXCL1, and CXCL2 between the gouty group and the healthy group, and we also predicted 10 chemicals related to these proteins. Conclusion: In this study, we screened and validated essential genes using a variety of bioinformatics tools to generate novel ideas for the diagnosis and treatment of gout.


Subject(s)
Biomarkers , Gene Expression Profiling , Gene Regulatory Networks , Gout , Humans , Gout/genetics , Chemokine CXCL1/genetics , Chemokine CXCL2/genetics , Chemokine CXCL2/metabolism , Computational Biology/methods , Transcriptome , THP-1 Cells , Interleukin-8/genetics , MicroRNAs/genetics , Uric Acid , Protein Interaction Maps , Gene Expression Regulation , Databases, Genetic , Arthritis, Gouty/genetics
7.
ACS Infect Dis ; 10(4): 1370-1378, 2024 04 12.
Article in English | MEDLINE | ID: mdl-38503263

ABSTRACT

Sepsis is a severe syndrome characterized by organ dysfunction, resulting from a systemic imbalance in response to infection. PAK1 plays a critical role in various diseases. The present study aimed to explore and delineate the mechanism of PAK1 in inflammation induced by sepsis. Bioinformatics analysis was performed to assess PAK1, snail, and CXCL2 expression in the whole blood of septic patients and the pathways enriched with PAK1. To simulate the sepsis model, THP-1 cells were stimulated with lipopolysaccharide. Gene expression was evaluated using qRT-PCR, while cell viability was assessed using CCK-8 assay. Cell apoptosis was tested with flow cytometry. Expression of inflammatory factors in cells following different treatments was analyzed using the enzyme linked immunosorbent assay (ELISA). Dual-luciferase and chromatin immunoprecipitation assays were conducted to verify the binding relationship between PAK1 and the snail. Mouse models of cecal ligation and puncture were established, and hematoxylin and eosin staining and ELISA were employed to detect the infiltration levels of inflammatory cells and the expression of related protective factors in lung, liver, and kidney tissues. The results demonstrated upregulation of PAK1, snail, and CXCL2 in the whole blood of septic patients, with PAK1 being enriched in the chemokine-related pathway. Knockdown of PAK1 significantly promoted the apoptosis of LPS-stimulated THP-1 cells and inhibited the expression of inflammatory factors. PAK1 upregulated the expression of the snail, which in turn promoted the expression of CXCL2. Thus, PAK1 mediated the sepsis-induced inflammatory response through the snail/CXCL2 pathway. In conclusion, PAK1 played a role in promoting inflammation induced by sepsis through the snail/CXCL2 axis, thereby providing a potential therapeutic target for the management of sepsis.


Subject(s)
Sepsis , Signal Transduction , Mice , Animals , Humans , Inflammation , Apoptosis , Liver/metabolism , Chemokine CXCL2/genetics , Chemokine CXCL2/metabolism , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
8.
Immunol Res ; 72(2): 212-224, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38351242

ABSTRACT

Acute restraint stress (RS) is routinely used to study the effects of psychological and/or physiological stress. We evaluated the impact of RS on cervical lymph nodes in rats at molecular and cellular levels. Male Sprague-Dawley rats were subjected to stress by immobilization for 30, 60, and 120 min (RS30, RS60, and RS120, respectively) and compared with rats of a no-stress control (C) group. The expression of genes encoding chemokines CXCL1/CXCL2 (Cxcl1 and Cxcl2) and their receptor CXCR2 (Cxcr2) was analyzed using reverse transcription-quantitative PCR (RT-qPCR) and microarray analyses. Immunohistochemistry and in situ hybridization were performed to determine the expression of these proteins and the macrophage biomarker CD68. Microarray analysis revealed that the expression of 514 and 496 genes was upregulated and downregulated, respectively, in the RS30 group. Compared with the C group, the RS30 group exhibited a 23.0-, 13.0-, and 1.6-fold increase in Cxcl1, Cxcl2, and Cxcr2 expression. Gene Ontology analysis revealed the involvement of these three upregulated genes in the cytokine network, inflammation, and leukocyte chemotaxis and migration. RT-qPCR analysis indicated that the mRNA levels of Cxcl1 and Cxcl2 were significantly increased in the RS30 group but were reverted to normal levels in the RS60 and RS120 groups. Cxcr2 mRNA level was significantly increased in the RS30 and RS120 groups compared with that in the C group. RS-induced CXCL1-immunopositive cells corresponded to B/plasma cells, whereas CXCL2-immunopositive cells corresponded to endothelial cells of the high endothelial venules. Stress-induced CXCR2-immunopositive cells corresponded to macrophages. Psychological and/or physiological stress induces an acute stress response and formation of an immunoreactive microenvironment in cervical lymph nodes, with the CXCL1/CXCL2-CXCR2 axis being pivotal in the acute stress response.


Subject(s)
Chemokines , Endothelial Cells , Rats , Male , Animals , Endothelial Cells/metabolism , Rats, Sprague-Dawley , Chemokines/genetics , Chemokine CXCL2/metabolism , Macrophages/metabolism , RNA, Messenger/genetics , Lymph Nodes/metabolism
9.
Biomed Chromatogr ; 38(5): e5837, 2024 May.
Article in English | MEDLINE | ID: mdl-38316604

ABSTRACT

Primary hepatocellular carcinoma (HCC) is one of the most common malignant tumors, but its pathogenesis remains incompletely elucidated. Recently, many studies indicated that lipid remodeling plays an important role in the occurrence and development of HCC. Furthermore, lipids have been proven to be indispensable mediators in promoting communication between tumor cells and extracellular matrix in the tumor microenvironment. Thus, this study aims to comprehensively investigate the process of lipid remodeling during HCC metastasis based on the LC-electrospray ionization-MS (LC-ESI-MS) combined with multiple reaction monitoring technology. M2 tumor-associated macrophages and the recombinant human protein CXCL2 were used to simulate the tumor microenvironment. After co-incubating SMMC7721 and MHCC97-H cell lines with M2 tumor-associated macrophages or the recombinant human protein CXCL2 for 48 h, LC-ESI-MS was used to quantify the levels of two major classes of lipid molecules, namely, glycerophospholipids and sphingolipids. Our results suggest that lipid remodeling in the tumor microenvironment may promote the migration and invasion of HCC cell lines.


Subject(s)
Carcinoma, Hepatocellular , Chemokine CXCL2 , Liver Neoplasms , Tumor-Associated Macrophages , Humans , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Cell Line, Tumor , Chemokine CXCL2/metabolism , Tumor-Associated Macrophages/metabolism , Lipid Metabolism , Tumor Microenvironment , Chromatography, Liquid/methods , Spectrometry, Mass, Electrospray Ionization/methods
10.
Adv Sci (Weinh) ; 10(35): e2304722, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37870186

ABSTRACT

Chronic low back pain (LBP) caused by intervertebral disc (IVD) degradation is a serious socioeconomic burden that can cause severe disabilities. Addressing the underlying pathogenic mechanisms of IVD degeneration may inspire novel therapeutic strategy for LBP. Herein, hypoxic preconditioning improves both the biological function of MSCs in hostile microenvironments and enhances the production of small extracellular vesicles (sEVs) with desirable therapeutic functions. In vitro results reveal that hypoxic preconditional engineering sEVs (HP-sEVs) alleviate the inflammatory microenvironments of IVD degradation, enhance the proliferation of nucleus pulposus (NP) cells, and promote proteoglycan synthesis and collagen formation. Transcriptomic sequencing reveales the excellent therapeutic effects of HP-sEVs in promoting extracellular matrix regeneration through the delivery of microRNA(miR)-7-5p, which further suppresses p65 production and thus the inhibition of Cxcl2 production. Moreover, in vivo results further confirm the robust therapeutic role of HP-sEVs in promoting IVD regeneration through the same mechanism mediated by miR-7-5p delivery. In conclusion, this study provides a novel therapeutic strategy for treating IVD degradation and is thus valuable for understanding the mechanism-of-action of HP-sEVs in IVD regeneration associated with chronic lower back pain.


Subject(s)
Extracellular Vesicles , Intervertebral Disc Degeneration , Intervertebral Disc , MicroRNAs , Humans , Extracellular Vesicles/metabolism , Intervertebral Disc/metabolism , Intervertebral Disc Degeneration/therapy , Intervertebral Disc Degeneration/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/metabolism , NF-kappa B/pharmacology , NF-kappa B/therapeutic use , Regeneration , Chemokine CXCL2/metabolism
11.
J Leukoc Biol ; 114(6): 666-671, 2023 11 24.
Article in English | MEDLINE | ID: mdl-37625009

ABSTRACT

Microbial infection is characterized by release of multiple proinflammatory chemokines that direct neutrophils to the insult site. How collective function of these chemokines orchestrates neutrophil recruitment is not known. Here, we characterized the role for heterodimer and show that the Cxcl1-Cxcl2 heterodimer is a potent neutrophil chemoattractant in mice and can recruit more neutrophils than the individual chemokines. Chemokine-mediated neutrophil recruitment is determined by Cxcr2 receptor signaling, Cxcr2 endocytosis, and binding to glycosaminoglycans. We have now determined heterodimer's Cxcr2 activity using cellular assays and Cxcr2 density in blood and recruited neutrophils in heterodimer-treated mice. We have shown that the heterodimer binds glycosaminoglycans with higher affinity and more efficiently than Cxcl1 or Cxcl2. These data collectively indicate that optimal glycosaminoglycan interactions and dampened receptor activity acting in concert in a dynamic fashion promote heterodimer-mediated robust neutrophil recruitment. We propose that this could play a critical role in combating infection.


Subject(s)
Chemokine CXCL1 , Chemokine CXCL2 , Neutrophils , Animals , Mice , Chemokine CXCL1/metabolism , Chemokine CXCL2/metabolism , Glycosaminoglycans/metabolism , Interleukin-8/metabolism , Neutrophil Infiltration , Neutrophils/metabolism , Receptors, Interleukin-8B/metabolism
12.
Cells ; 12(14)2023 07 09.
Article in English | MEDLINE | ID: mdl-37508478

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a multifactorial neurodegenerative disease characterized by progressive depletion of motor neurons (MNs). Recent evidence suggests a role in ALS pathology for the C-X-C motif chemokine receptor 2 (CXCR2), whose expression was found increased at both mRNA and protein level in cortical neurons of sporadic ALS patients. Previous findings also showed that the receptor inhibition is able to prevent iPSC-derived MNs degeneration in vitro and improve neuromuscular function in SOD1-G93A mice. Here, by performing transcriptional analysis and immunofluorescence studies, we detailed the increased expression and localization of CXCR2 and its main ligand CXCL8 in the human lumbar spinal cord of sporadic ALS patients. We further investigated the functional role of CXCR2/ligands axis in NSC-34 motor neuron-like cells expressing human wild-type (WT) or mutant (G93A) SOD1. A significant expression of CXCR2 was found in doxycycline-induced G93A-SOD1-expressing cells, but not in WT cells. In vitro assays showed CXCR2 activation by GROα and MIP2α, two murine endogenous ligands and functional homologs of CXCL8, reduces cellular viability and triggers apoptosis in a dose dependent manner, while treatment with reparixin, a non-competitive allosteric CXCR2 inhibitor, effectively counteracts GROα and MIP2α toxicity, significantly inhibiting the chemokine-induced cell death. Altogether, data further support a role of CXCR2 axis in ALS etiopathogenesis and confirm its pharmacological modulation as a candidate therapeutic strategy.


Subject(s)
Amyotrophic Lateral Sclerosis , Neurodegenerative Diseases , Animals , Humans , Mice , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Apoptosis , Chemokine CXCL2/metabolism , Ligands , Mice, Transgenic , Motor Neurons/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism
13.
Nat Commun ; 14(1): 1513, 2023 03 18.
Article in English | MEDLINE | ID: mdl-36934105

ABSTRACT

Hepatocytes function largely through the secretion of proteins that regulate cell proliferation, metabolism, and intercellular communications. During the progression of hepatocellular carcinoma (HCC), the hepatocyte secretome changes dynamically as both a consequence and a causative factor in tumorigenesis, although the full scope of secreted protein function in this process remains unclear. Here, we show that the secreted pseudo serine protease PRSS35 functions as a tumor suppressor in HCC. Mechanistically, we demonstrate that active PRSS35 is processed via cleavage by proprotein convertases. Active PRSS35 then suppresses protein levels of CXCL2 through targeted cleavage of tandem lysine (KK) recognition motif. Consequently, CXCL2 degradation attenuates neutrophil recruitment to tumors and formation of neutrophil extracellular traps, ultimately suppressing HCC progression. These findings expand our understanding of the hepatocyte secretome's role in cancer development while providing a basis for the clinical translation of PRRS35 as a therapeutic target or diagnostic biomarker.


Subject(s)
Carcinoma, Hepatocellular , Extracellular Traps , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Extracellular Traps/metabolism , Peptide Hydrolases/metabolism , Hepatocytes/metabolism , Cell Line, Tumor , Chemokine CXCL2/metabolism
14.
Infect Immun ; 91(4): e0001423, 2023 04 18.
Article in English | MEDLINE | ID: mdl-36880752

ABSTRACT

Staphylococcus aureus is the principal causative agent of osteomyelitis, a serious bacterial infection of bone that is associated with progressive inflammatory damage. Bone-forming osteoblasts have increasingly been recognized to play an important role in the initiation and progression of detrimental inflammation at sites of infection and have been demonstrated to release an array of inflammatory mediators and factors that promote osteoclastogenesis and leukocyte recruitment following bacterial challenge. In the present study, we describe elevated bone tissue levels of the potent neutrophil-attracting chemokines CXCL1, CXCL2, CXCL3, CXCL5, CCL3, and CCL7 in a murine model of posttraumatic staphylococcal osteomyelitis. RNA sequencing (RNA-Seq) gene ontology analysis of isolated primary murine osteoblasts showed enrichment in differentially expressed genes involved in cell migration and chemokine receptor binding and chemokine activity following S. aureus infection, and a rapid increase in the expression of mRNA encoding CXCL1, CXCL2, CXCL3, CXCL5, CCL3, and CCL7, in these cells. Importantly, we have confirmed that such upregulated gene expression results in protein production with the demonstration that S. aureus challenge elicits the rapid and robust release of these chemokines by osteoblasts and does so in a bacterial dose-dependent manner. Furthermore, we have confirmed the ability of soluble osteoblast-derived chemokines to elicit the migration of a neutrophil-like cell line. As such, these studies demonstrate the robust production of CXCL1, CXCL2, CXCL3, CXCL5, CCL3, and CCL7 by osteoblasts in response to S. aureus infection, and the release of such neutrophil-attracting chemokines provides an additional mechanism by which osteoblasts could drive the inflammatory bone loss associated with staphylococcal osteomyelitis.


Subject(s)
Osteomyelitis , Staphylococcal Infections , Animals , Mice , Staphylococcus aureus/metabolism , Neutrophils/metabolism , Chemokines/metabolism , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Osteoblasts , Interleukin-8/metabolism , Staphylococcal Infections/microbiology , Chemokine CXCL2/genetics , Chemokine CXCL2/metabolism , Chemokine CCL7/metabolism , Chemokine CCL3/metabolism
15.
Phytomedicine ; 111: 154662, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36681054

ABSTRACT

BACKGROUND: Postoperative abdominal adhesion (PAA) is the most common complication after abdominal surgeries, which can lead to intestinal obstruction, chronic abdominal pain or female infertility. Jiawei Xiaochengqi decoction (JWXCQ) is a hospital preparation widely used for PAA treatment in Nanfang Hospital of Southern Medical University for more than twenty years. PURPOSE: This study aimed to investigate the therapeutic effects and potential mechanism of JWXCQ against PAA and provide beneficial information for its clinical application. METHODS: The main active components of JWXCQ were identified using ultra high performance liquid chromatography (UHPLC) combined with standard substance comparison. The efficacy and underlying mechanism of JWXCQ were evaluated through in vivo experiments with a postsurgical-induced peritoneal adhesion rat model, and in vitro studies with LPS-stimulated Raw 264.7 macrophages and primary fibroblasts. H&E and Masson staining were performed to assess histopathological changes. The levels of cytokines/proteins-associated with inflammation and degradation of extracellular matrix as well as CXCL2-CXCR2 pathway-related proteins were determined by ELISA, qRT-PCR, western blot assays or immunohistochemistry, respectively. Furthermore, siCXCR2 transfection was used to validate the mechanism of action of JWXCQ. RESULTS: JWXCQ treatment significantly reduced the formation of PAA, inhibited the inflammation and collagen deposition, and facilitated the secretion of MMP9, decreased the levels of IL-1ß, IL-6, TIMP1, COL-1, and suppressed the CXCL2-CXCR2 pathway in PAA rats. Furthermore, JWXCQ inhibited its downstream pathways, the JAK2-STAT3 and PI3K-AKT signaling, as indicated by the suppression of the phosphorylation levels of STAT3 and AKT. In vitro cell experiments revealed that JWXCQ reduced IL-1ß and IL-6 secretion in Raw 264.7 macrophages and COL-1 in primary fibroblasts. The CXCL2-CXCR2, JAK2-STAT3 and PI3K-AKT pathways were also inhibited after JWXCQ treatment, which were consistent with the in vivo results. More importantly, silence of CXCR2 eliminated the regulatory effects of JWXCQ. CONCLUSION: JWXCQ could effectively prevent the PAA formation by alleviating inflammation and collagen deposition, which was associated with the inhibition of CXCL2-CXCR2 pathway. This study investigated the relevant pharmacological mechanisms of JWXCQ, providing further evidence for the application of JWXCQ in clinical PAA treatment.


Subject(s)
Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Animals , Female , Rats , Chemokine CXCL2/metabolism , Cytokines/metabolism , Inflammation/drug therapy , Interleukin-6
16.
Biochim Biophys Acta Mol Basis Dis ; 1868(12): 166490, 2022 12 01.
Article in English | MEDLINE | ID: mdl-35841983

ABSTRACT

BACKGROUND: Acute pancreatitis (AP) was initiated within pancreatic parenchymal cells and sustained by uncontrolled inflammatory responses. AXL and MERTK receptor tyrosine kinases play a crucial role in negatively regulating the innate immunity. Therefore, this study aimed to investigate the role and underlying mechanism of AXL and MERTK in AP. METHODS: Experimental AP was induced by ten hourly intraperitoneal administration of caerulein in global, hematopoietic- and pancreas-specific Axl and Mertk deficient mice. Pancreatitis severity was assessed biochemically and histologically. Pancreatic transcriptomics and pancreatic infiltrating immune cells were profiled. Some mice were given R428, an antagonist of AXL and MERTK. AXL and MERTK in peripheral leukocytes were measured by flow cytometry. FINDINGS: The levels of AXL and MERTK in pancreatic tissue and pancreatic CD45+ cells were dynamically altered at 6 h and 12 h after the 1st injection of caerulein. Global and hematopoietic-specific, but not pancreas-specific deletion of Axl and Mertk protected against pancreatic necrosis and trypsinogen activation. Pancreatic transcriptomic analysis revealed that differentially expressed gene signatures were mainly related to metabolic and inflammatory pathways. Furthermore, deletion or inhibition of Axl and Mertk selectively inhibited pancreatic neutrophil infiltration, which was primarily related to CXCL2 secreted by pro-inflammatory macrophages. Increased levels of MERTK in peripheral leukocytes were correlated with more severe form of AP. INTERPRETATION: Our findings reveal that specific AXL/MERTK antagonist may be a novel and potential early treatment for AP and the levels of MERTK in peripheral leukocytes may be a promising biomarker for predicting pancreatic severity in patients with AP. FUNDING: National Natural Science Foundation of China, Shanghai Natural Science Foundation, a Shanghai Young Talent Award and a Shanghai Young Orient Scholar Award. RESEARCH IN CONTEXT: Evidence before this study Acute pancreatitis (AP) is a common inflammatory disorder of the exocrine pancreas, the severity of which was determined by the extent of pancreatic necrosis, with no targeted therapy. AP was initiated by signals within pancreatic parenchymal cells and sustained by uncontrolled innate immune responses. One of the three crucial regulatory roles for AXL and MERTK is to negatively regulate innate immune responses. Added value of this study Global and hematopoietic-, but not pancreas-specific Axl and Mertk deficiency protected against pancreatitis, primarily pancreatic necrosis. Deletion of Axl and Mertk selectively inhibited pancreatic neutrophil infiltration that was related to CXCL2 secreted by pro-inflammatory macrophages. AXL and MERTK antagonist similarly reduced pancreatitis severity via limiting CXCL2-mediated pancreatic neutrophil infiltration. Higher levels of MERTK, but not AXL in peripheral leukocytes were correlated with more severe form of acute pancreatitis. Implications of all the available evidence A specific AXL/MERTK antagonist may be a novel and potential early treatment for AP. The level of MERTK on peripheral leukocytes may be a promising biomarker for predicting disease severity in patients with AP.


Subject(s)
Ceruletide , Pancreatitis, Acute Necrotizing , Acute Disease , Animals , Chemokine CXCL2/metabolism , China , Mice , Mice, Inbred C57BL , Neutrophil Infiltration , Trypsinogen/metabolism , Tyrosine , c-Mer Tyrosine Kinase/genetics
17.
Int Heart J ; 63(4): 755-762, 2022 Jul 30.
Article in English | MEDLINE | ID: mdl-35831147

ABSTRACT

Ursolic acid (UA) has been reported to possess several biological benefits, such as anti-cancer, anti-inflammation, antibacterial, and neuroprotective functions. This study detects the function and molecular mechanism of UA in H9c2 cells under hypoxia and reoxygenation (H/R) conditions.Under H/R stimulation, the effects of UA on H9c2 cells were examined using ELISA and western blot assays. The Comparative Toxicogenomics Database was employed to analyze the target molecule of UA. Small interfering RNA was used to knock down CXCL2 expression, further exploring the function of CXCL2 in H/R-induced H9c2 cells. The genes related to the nuclear factor-kappa B (NF-κB) pathway were assessed using western blot analysis.Significant effects of UA on H/R-induced H9c2 cell damage were observed, accompanied by reduced inflammation and oxidative stress injury. Additionally, the increased level of CXCL2 in H/R-induced H9c2 cells was reduced after UA stimulation. Moreover, CXCL2 knockdown strengthened the beneficial effect of UA on H/R-induced H9c2 cells. HY-18739, an activator of the NF-κB pathway, can increase CXCL2 expression. Moreover, the increased levels of p-P65 NF-κB and p-IκBα in H/R-induced H9c2 cells were remarkably attenuated by UA treatment.In summary, the results indicated that UA may alleviate the damage of H9c2 cells by targeting the CXCL2/NF-κB pathway under H/R conditions.


Subject(s)
NF-kappa B , Triterpenes , Anti-Inflammatory Agents/pharmacology , Apoptosis , Chemokine CXCL2/metabolism , Chemokine CXCL2/pharmacology , Humans , Hypoxia/metabolism , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Signal Transduction , Triterpenes/metabolism , Triterpenes/pharmacology , Ursolic Acid
18.
Sci Rep ; 12(1): 486, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35017565

ABSTRACT

Uropathogenic Escherichia coli (UPEC) may undergo a cyclic cascade of morphological alterations that are believed to enhance the potential of UPEC to evade host responses and re-infect host cell. However, knowledge on the pathogenic potential and host activation properties of UPEC during the morphological switch is limited. Microarray analysis was performed on mRNA isolated from human bladder epithelial cells (HBEP) after exposure to three different morphological states of UPEC (normal coliform, filamentous form and reverted form). Cells stimulated with filamentous bacteria showed the lowest number of significant gene alterations, although the number of enriched gene ontology classes was high suggesting diverse effects on many different classes of host genes. The normal coliform was in general superior in stimulating transcriptional activity in HBEP cells compared to the filamentous and reverted form. Top-scored gene entities activated by all three morphological states included IL17C, TNFAIP6, TNF, IL20, CXCL2, CXCL3, IL6 and CXCL8. The number of significantly changed canonical pathways was lower in HBEP cells stimulated with the reverted form (32 pathways), than in cells stimulated with the coliform (83 pathways) or filamentous bacteria (138 pathways). A host cell invasion assay showed that filamentous bacteria were unable to invade bladder cells, and that the number of intracellular bacteria was markedly lower in cells infected with the reverted form compared to the coliform. In conclusion, the morphological state of UPEC has major impact on the host bladder response both when evaluating the number and the identity of altered host genes and pathways.


Subject(s)
Epithelial Cells/metabolism , Escherichia coli Infections/genetics , Transcription, Genetic , Urinary Bladder/microbiology , Uropathogenic Escherichia coli/physiology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Line , Chemokine CXCL2/genetics , Chemokine CXCL2/metabolism , Chemokines, CXC/genetics , Chemokines, CXC/metabolism , Epithelial Cells/microbiology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Urinary Bladder/metabolism , Uropathogenic Escherichia coli/growth & development
19.
Exp Eye Res ; 216: 108943, 2022 03.
Article in English | MEDLINE | ID: mdl-35074346

ABSTRACT

Fibroblast-stimulating lipopeptide (FSL-1) can activate Toll-like receptor 2 and 6 (TLR2/6), which recognize relevant molecules from gram-positive pathogens, fungus, and mycoplasma, and elevates the expression of CXCL1 and CXCL2, neutrophil chemoattractants, in certain types of cells. This effect has not previously been reported in the uveal melanocytes (UM). This study was designed to test the hypothesis that FSL-1 can induce the expression and secretion of CXCL1 and CXCL2 via activation of TLR2/6 in cultured human UM and producing an acute non-infectious uveitis reaction in the mouse. Flow cytometry and fluorescent immunostaining were used to measure the effect of FSL-1 on the expression of TLR2/6 in UM. Real time PCR and ELISA analysis were used to assess the ability of FSL-1 to elevate CXCL1/CXCL2 levels in cell lysates and conditioned media of UM, respectively. Flow cytometry measured phosphorylated MAPK and activated NF-κB signals in UM, with and without FSL-1 treatment. ELISA analysis tested the impact of various signal inhibitors (NF-κB, p38 MAPK, JNK1/2 and ERK1/2) and TLR2/6 antagonists on FSL-1-induced CXCL1/CXCL2 levels in cultured UM. The effects of neutralizing antibodies to TLR2 on FSL-1-induced mouse uveitis were tested in an experimental animal model. FSL-1 induced the expression of TLR2/6 proteins in cultured UM. FSL-1 significantly elevated the CXCL1 and CXCL2 proteins and mRNA levels in cultured UM time- and dose-dependently. FSL-1 mainly activated NF-κB, JNK, and expression of TLR2. FSL-1-induced expression of CXCL1 and CXCL2 was blocked by NF-κB, JNK, ERK inhibitors and TLR2 antagonists. Intravitreal injection of FSL-1 induced acute non-infectious mouse uveitis, which was significantly reduced in severity by a TLR2 antagonist. These results suggest that UM may play a role in the immune reaction, which targets invading pathogens, especially gram-positive bacteria. On the other hand, an excessive reaction to molecules from gram-positive bacteria may promote an inflammatory state of non-infectious uveitis.


Subject(s)
Chemokine CXCL1/metabolism , Chemokine CXCL2/metabolism , Diglycerides/pharmacology , Melanocytes/drug effects , Oligopeptides/pharmacology , Toll-Like Receptor 2/agonists , Toll-Like Receptor 6/agonists , Uvea/cytology , Animals , Antibodies, Neutralizing/pharmacology , Cells, Cultured , Chemokine CXCL1/genetics , Chemokine CXCL2/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Humans , Intravitreal Injections , Melanocytes/metabolism , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/metabolism , Phosphorylation , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Uveitis/chemically induced , Uveitis/metabolism
20.
Int J Mol Sci ; 24(1)2022 Dec 22.
Article in English | MEDLINE | ID: mdl-36613652

ABSTRACT

In this paper, we present a literature review of the role of CXC motif chemokine ligand 1 (CXCL1) in physiology, and in selected major non-cancer diseases of the cardiovascular system, respiratory system and skin. CXCL1, a cytokine belonging to the CXC sub-family of chemokines with CXC motif chemokine receptor 2 (CXCR2) as its main receptor, causes the migration and infiltration of neutrophils to the sites of high expression. This implicates CXCL1 in many adverse conditions associated with inflammation and the accumulation of neutrophils. The aim of this study was to describe the significance of CXCL1 in selected diseases of the cardiovascular system (atherosclerosis, atrial fibrillation, chronic ischemic heart disease, hypertension, sepsis including sepsis-associated encephalopathy and sepsis-associated acute kidney injury), the respiratory system (asthma, chronic obstructive pulmonary disease (COPD), chronic rhinosinusitis, coronavirus disease 2019 (COVID-19), influenza, lung transplantation and ischemic-reperfusion injury and tuberculosis) and the skin (wound healing, psoriasis, sunburn and xeroderma pigmentosum). Additionally, the significance of CXCL1 is described in vascular physiology, such as the effects of CXCL1 on angiogenesis and arteriogenesis.


Subject(s)
Cardiovascular Diseases , Chemokine CXCL1 , Respiratory Tract Diseases , Skin Diseases , Humans , Cardiovascular System/metabolism , Chemokine CXCL1/metabolism , Chemokine CXCL2/metabolism , Chemokines/metabolism , Lung/metabolism , Neutrophil Infiltration , Neutrophils/metabolism , Receptors, Interleukin-8B/metabolism , Respiratory System , Skin
SELECTION OF CITATIONS
SEARCH DETAIL