Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.188
Filter
1.
Arh Hig Rada Toksikol ; 75(2): 102-109, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38963144

ABSTRACT

COVID-19 can cause a range of complications, including cardiovascular, renal, and/or respiratory insufficiencies, yet little is known of its potential effects in persons exposed to toxic metals. The aim of this study was to answer this question with in silico toxicogenomic methods that can provide molecular insights into COVID-19 complications owed to exposure to arsenic, cadmium, lead, mercury, nickel, and chromium. For this purpose we relied on the Comparative Toxicogenomic Database (CTD), GeneMANIA, and ToppGene Suite portal and identified a set of five common genes (IL1B, CXCL8, IL6, IL10, TNF) for the six metals and COVID-19, all of which code for pro-inflammatory and anti-inflammatory cytokines. The list was expanded with additional 20 related genes. Physical interactions are the most common between the genes affected by the six metals (77.64 %), while the dominant interaction between the genes affected by each metal separately is co-expression (As 56.35 %, Cd 64.07 %, Pb 71.5 %, Hg 81.91 %, Ni 64.28 %, Cr 88.51 %). Biological processes, molecular functions, and pathways in which these 25 genes participate are closely related to cytokines and cytokine storm implicated in the development of COVID-19 complications. In other words, our findings confirm that exposure to toxic metals, alone or in combinations, might escalate COVID-19 severity.


Subject(s)
COVID-19 , Cadmium , Mercury , Humans , Cadmium/toxicity , Mercury/toxicity , Lead/toxicity , Computer Simulation , SARS-CoV-2 , Arsenic/toxicity , Nickel/toxicity , Metals, Heavy/toxicity , Chromium/toxicity , Cytokines , Interleukin-1beta/genetics , Interleukin-8/genetics , Toxicogenetics , Interleukin-6/genetics , Interleukin-10/genetics , Tumor Necrosis Factor-alpha/genetics
2.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000270

ABSTRACT

The combination of a polyphenol, quercetin, with dasatinib initiated clinical trials to evaluate the safety and efficacy of senolytics in idiopathic pulmonary fibrosis, a lung disease associated with the presence of senescent cells. Another approach to senotherapeutics consists of controlling inflammation related to cellular senescence or "inflammaging", which participates, among other processes, in establishing pulmonary fibrosis. We evaluate whether polyphenols such as caffeic acid, chlorogenic acid, epicatechin, gallic acid, quercetin, or resveratrol combined with different senotherapeutics such as metformin or rapamycin, and antifibrotic drugs such as nintedanib or pirfenidone, could present beneficial actions in an in vitro model of senescent MRC-5 lung fibroblasts. A senescent-associated secretory phenotype (SASP) was evaluated by the measurement of interleukin (IL)-6, IL-8, and IL-1ß. The senescent-associated ß-galactosidase (SA-ß-gal) activity and cellular proliferation were assessed. Fibrosis was evaluated using a Picrosirius red assay and the gene expression of fibrosis-related genes. Epithelial-mesenchymal transition (EMT) was assayed in the A549 cell line exposed to Transforming Growth Factor (TGF)-ß in vitro. The combination that demonstrated the best results was metformin and caffeic acid, by inhibiting IL-6 and IL-8 in senescent MRC-5 cells. Metformin and caffeic acid also restore cellular proliferation and reduce SA-ß-gal activity during senescence induction. The collagen production by senescent MRC-5 cells was inhibited by epicatechin alone or combined with drugs. Epicatechin and nintedanib were able to control EMT in A549 cells. In conclusion, caffeic acid and epicatechin can potentially increase the effectiveness of senotherapeutic drugs in controlling lung diseases whose pathophysiological component is the presence of senescent cells and fibrosis.


Subject(s)
Cellular Senescence , Fibroblasts , Lung , Polyphenols , Humans , Fibroblasts/drug effects , Fibroblasts/metabolism , Cellular Senescence/drug effects , Polyphenols/pharmacology , Lung/pathology , Lung/drug effects , Lung/metabolism , A549 Cells , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Metformin/pharmacology , Caffeic Acids/pharmacology , Indoles/pharmacology , Senotherapeutics/pharmacology , Cell Line , Senescence-Associated Secretory Phenotype/drug effects , Sirolimus/pharmacology , Interleukin-8/metabolism , Interleukin-8/genetics , Transforming Growth Factor beta/metabolism , Pyridones
3.
Sci Rep ; 14(1): 16471, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39014096

ABSTRACT

Psoriasis, a chronic inflammatory skin disorder, is associated with comorbidities such as acute myocardial infarction (AMI). However, the molecular mechanisms connecting these conditions are unclear. In this study, we conducted bioinformatics analyses using gene expression datasets to identify differentially expressed genes and hub genes associated with both psoriasis and AMI. Our findings emphasize the involvement of immune-related pathways in the pathogenesis of both conditions. Furthermore, we investigated the expression levels of hub genes in AMI patients and myocardial infarction (MI) mice. ELISA measurements revealed significantly higher levels of CXCL8, IL1B, S100A9, and S100A12 in the serum of AMI patients compared to normal individuals. Immunohistochemical staining of heart tissue from MI mice showed a progressive increase in the expression of CXCL8 and IL-1B as MI advanced, while S100A9 exhibited high expression at day 3 post-MI. mRNA expression analysis validated these findings. Additionally, we explored the skin lesions of psoriasis patients and found significantly higher expression of CXCL8, IL-1B, S100A9, and S100A12 in the affected skin areas compared to unaffected regions. These results highlight the consistent upregulation of hub genes in both AMI and psoriasis patients, as well as in myocardial infarction mice, underscoring their potential as reliable markers for disease diagnosis. Moreover, molecular docking simulations revealed potential interactions between simvastatin and key target proteins, suggesting a potential therapeutic avenue. Overall, our study uncovers shared molecular signatures and potential therapeutic targets, providing a foundation for future investigations targeting common pathways in psoriasis and AMI.


Subject(s)
Calgranulin B , Myocardial Infarction , Psoriasis , Psoriasis/genetics , Psoriasis/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Animals , Humans , Mice , Calgranulin B/genetics , Calgranulin B/metabolism , Interleukin-8/metabolism , Interleukin-8/genetics , Molecular Docking Simulation , Simvastatin/pharmacology , Simvastatin/therapeutic use , S100A12 Protein/genetics , S100A12 Protein/metabolism , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Male , Disease Models, Animal , Computational Biology/methods , Gene Expression Profiling , Female , Biomarkers
4.
Redox Biol ; 74: 103209, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38861833

ABSTRACT

Alterations in the tumor microenvironment are closely associated with the metabolic phenotype of tumor cells. Cancer-associated fibroblasts (CAFs) play a pivotal role in tumor growth and metastasis. Existing studies have suggested that lactate produced by tumor cells can activate CAFs, yet the precise underlying mechanisms remain largely unexplored. In this study, we initially identified that lactate derived from lung cancer cells can promote nuclear translocation of NUSAP1, subsequently leading to the recruitment of the transcriptional complex JUNB-FRA1-FRA2 near the DESMIN promoter and facilitating DESMIN transcriptional activation, thereby promoting CAFs' activation. Moreover, DESMIN-positive CAFs, in turn, secrete IL-8, which recruits TAMs or promotes M2 polarization of macrophages, further contributing to the alterations in the tumor microenvironment and facilitating lung cancer progression. Furthermore, we observed that the use of IL-8 receptor antagonists, SB225002, or Navarixin, significantly reduced TAM infiltration and enhanced the therapeutic efficacy of anti-PD-1 or anti-PD-L1 treatment. This finding indicates that inhibiting IL-8R activity can attenuate the impact of CAFs on the tumor microenvironment, thus restraining the progression of lung cancer.


Subject(s)
Cancer-Associated Fibroblasts , Interleukin-8 , Lung Neoplasms , Macrophages , Tumor Microenvironment , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Humans , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Interleukin-8/metabolism , Interleukin-8/genetics , Mice , Animals , Macrophages/metabolism , Macrophages/immunology , Lactic Acid/metabolism , Disease Progression , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology
5.
Anim Sci J ; 95(1): e13972, 2024.
Article in English | MEDLINE | ID: mdl-38923622

ABSTRACT

High grain feeding or weaning, which could compromise the rumen epithelium by increasing ruminal short-chain fatty acid (SCFA) concentrations with pH reduction, is associated with high levels of ruminal toll-like receptor 5 (TLR5). This study aimed to determine the role of TLR5 in the rumen epithelium. Immunohistochemistry revealed that TLR5 was localized in cells on the basal side (i.e., basal and spinous layers) rather than in the granular layer in the rumen epithelium, where tight junctions are most potent, in pre- and post-weaning calves (n = 9). Primary bovine rumen epithelial cells (BRECs) obtained from Holstein cows (n = 3) were cultured to investigate the factors that upregulate TLR5; however, SCFA, low pH (pH 5.6), BHBA, L-lactate, D-lactate, and LPS did not upregulate TLR5 gene expression in BREC. Primary BREC treated with flagellin (TLR5 ligand) had higher expression of interleukin-1ß (IL-1ß) (P < 0.05) than BREC treated with vehicle. In addition, BREC treated with IL-1ß had higher expression of antimicrobial peptides and C-X-C motif chemokine ligand 8 than BREC treated with vehicle (P < 0.05). These results suggest that ruminal TLR5 may recognize epithelial disruption via flagellin and mediate the immune response via IL-1ß during high-grain feeding or weaning.


Subject(s)
Epithelial Cells , Gene Expression , Interleukin-1beta , Interleukin-8 , Rumen , Toll-Like Receptor 5 , Animals , Toll-Like Receptor 5/genetics , Toll-Like Receptor 5/metabolism , Rumen/metabolism , Cattle/metabolism , Epithelial Cells/metabolism , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Cells, Cultured , Interleukin-8/metabolism , Interleukin-8/genetics , Weaning , Antimicrobial Peptides/genetics , Antimicrobial Peptides/metabolism , Flagellin/pharmacology , Fatty Acids, Volatile/metabolism , Fatty Acids, Volatile/pharmacology , Ligands , Up-Regulation
6.
Biomed Khim ; 70(3): 156-160, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38940204

ABSTRACT

The cytokine profile of primary coronary artery endothelial cells cultivated in the presence of doxorubicin (2 µg/ml and 6 µg/ml) was evaluated using enzyme-linked immunosorbent assay and qPCR. Cultivation of cells in the presence of these concentrations of doxorubicin for 24 h, upregulated expression of the following genes: IL6 (by 2.30 and 2.66 times, respectively), IL1B (by 1.25 and 3.44 times), and CXCL8 (by 6.47 times and 6.42 times), MIF (2.34 and 2.28 times), CCL2 (4.22 and 3.98 times). Under these conditions the following genes were downregulated: IL10, IL1R2, TNF. Cultivation of cells in the presence of doxorubicin (2 µg/ml and 6 µg/ml) for 24 h also increased the secretion of IL-6.


Subject(s)
Coronary Vessels , Doxorubicin , Endothelial Cells , Interleukin-6 , Humans , Doxorubicin/pharmacology , Coronary Vessels/cytology , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Interleukin-6/metabolism , Interleukin-6/genetics , Cells, Cultured , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Cytokines/metabolism , Cytokines/genetics , Gene Expression Regulation/drug effects , Interleukin-8/metabolism , Interleukin-8/genetics , Chemokine CCL2/metabolism , Chemokine CCL2/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Interleukin-10/metabolism , Interleukin-10/genetics
7.
Int J Mol Sci ; 25(11)2024 May 23.
Article in English | MEDLINE | ID: mdl-38891846

ABSTRACT

Tumor recurrence and drug resistance are responsible for poor prognosis in colorectal cancer (CRC). DNA mismatch repair (MMR) deficiency or elevated interleukin-8 (IL-8) levels are characteristics of CRCs, which have been independently correlated with treatment resistance to common therapies. We recently demonstrated significantly impaired therapeutical response and increased IL-8 release of CRC cell lines with reduced expression of MMR protein MLH1 as well as cytoskeletal non-erythrocytic spectrin alpha II (SPTAN1). In the present study, decreased intratumoral MLH1 and SPTAN1 expression in CRCs could be significantly correlated with enhanced serum IL-8. Furthermore, using stably reduced SPTAN1-expressing SW480, SW620 or HT-29 cell lines, the RAS-mediated RAF/MEK/ERK pathway was analyzed. Here, a close connection between low SPTAN1 expression, increased IL-8 secretion, enhanced extracellular-signal-regulated kinase (ERK) phosphorylation and a mesenchymal phenotype were detected. The inhibition of ERK by U0126 led to a significant reduction in IL-8 secretion, and the combination therapy of U0126 with FOLFOX optimizes the response of corresponding cancer cell lines. Therefore, we hypothesize that the combination therapy of FOLFOX and U0126 may have great potential to improve drug efficacy on this subgroup of CRCs, showing decreased MLH1 and SPTAN1 accompanied with high serum IL-8 in affected patients.


Subject(s)
Butadienes , Colorectal Neoplasms , Fluorouracil , Interleukin-8 , Nitriles , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Interleukin-8/metabolism , Interleukin-8/genetics , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Butadienes/pharmacology , Nitriles/pharmacology , Cell Line, Tumor , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/therapeutic use , Leucovorin/therapeutic use , Leucovorin/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Female , Male , Extracellular Signal-Regulated MAP Kinases/metabolism , HT29 Cells , MAP Kinase Signaling System/drug effects , MutL Protein Homolog 1/metabolism , MutL Protein Homolog 1/genetics , Middle Aged , Aged , Gene Expression Regulation, Neoplastic/drug effects , Phosphorylation/drug effects
8.
Br J Cancer ; 131(2): 271-282, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38862740

ABSTRACT

BACKGROUND: Cancer-associated fibroblasts (CAFs)-derived IL-8 plays important roles in chemoresistance, immunosuppression, and lymph node metastasis of gastric cancer. However, the mechanisms underlying IL-8 production in CAFs remains unclear. METHODS: DNA pulldown assay was performed to identify the transcription factors responsible for IL-8 expression in CAFs, which was further verified using CHIP-qPCR and DNA agarose gel electrophoresis assays. The cellular localisation of IL-8 was analysed using multiplex immunofluorescence (MxIF). RESULTS: MxIF demonstrated that IL-8 was mainly produced by CAFs in gastric cancer. Lysine[K]-specific demethylase 5B (KDM5B) was identified as an IL-8 transcription factor in CAFs, and the binding of KDM5B to phosphorylated RB1 limited the transcriptional regulation of IL-8 in gastric cancer cells. Serglycin (SRGN) secreted by tumour cells activated the CD44/c-Myc pathway to upregulate KDM5B expression, thereby promoting IL-8 production in CAFs. Furthermore, tumour-associated neutrophils (TANs)-derived regenerating family member 4 (REG4) upregulates SRGN expression by activating cAMP-responsive element binding protein 1 (CREB1) in gastric cancer cells. Thus, the SRGN-IL-8-TANs-SRGN loop, which facilitates tumour progression, has been explored in gastric cancer. CONCLUSIONS: This study revealed the mechanisms of the preferential production of IL-8 by CAFs in gastric cancer, and paves the way for potential new therapeutic strategies for gastric cancer.


Subject(s)
Cancer-Associated Fibroblasts , Interleukin-8 , Proteoglycans , Stomach Neoplasms , Vesicular Transport Proteins , Humans , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Interleukin-8/metabolism , Interleukin-8/genetics , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Proteoglycans/metabolism , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Cell Line, Tumor , Jumonji Domain-Containing Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Gene Expression Regulation, Neoplastic , Cyclic AMP Response Element-Binding Protein/metabolism , Animals , Mice , Male
9.
Fish Shellfish Immunol ; 150: 109647, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38797335

ABSTRACT

NIK (NF-κB inducing kinase) belongs to the mitogen-activated protein kinase family, which activates NF-κB and plays a vital role in immunology, inflammation, apoptosis, and a series of pathological responses. In NF-κB noncanonical pathway, NIK and IKKα have been often studied in mammals and zebrafish. However, few have explored the relationship between NIK and other subunits of the IKK complex. As a classic kinase in the NF-κB canonical pathway, IKKß has never been researched with NIK in fish. In this paper, the full-length cDNA sequence of grass carp (Ctenopharyngodon idella) NIK (CiNIK) was first cloned and identified. The expression level of CiNIK in grass carp cells was increased under GCRV stimuli. Under the stimulation of GCRV, poly (I:C), and LPS, the expression of NIK in various tissues of grass carp was also increased. This suggests that CiNIK responds to viral stimuli. To study the relationship between CiNIK and CiIKKß, we co-transfected CiNIK-FLAG and CiIKKB-GFP into grass carp cells in coimmunoprecipitation and immunofluorescence experiments. The results revealed that CiNIK interacts with CiIKKß. Besides, the degree of autophosphorylation of CiNIK was enhanced under poly (I:C) stimulation. CiIKKß was phosphorylated by CiNIK and then activated the activity of p65. The activity change of p65 indicates that NF-κB downstream inflammatory genes will be functioning. CiNIK or CiIKKß up-regulated the expression of IL-8. It got higher when CiNIK and CiIKKß coexisted. This paper revealed that NF-κB canonical pathway and noncanonical pathway are not completely separated in generating benefits.


Subject(s)
Amino Acid Sequence , Carps , Fish Proteins , Interleukin-8 , NF-kappa B , Protein Serine-Threonine Kinases , Up-Regulation , Animals , Carps/genetics , Carps/immunology , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/chemistry , NF-kappa B/genetics , NF-kappa B/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Interleukin-8/immunology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Fish Diseases/immunology , Signal Transduction , Reoviridae/physiology , Phylogeny , NF-kappaB-Inducing Kinase , Gene Expression Regulation/immunology , Poly I-C/pharmacology , Lipopolysaccharides/pharmacology , Reoviridae Infections/immunology , Reoviridae Infections/veterinary , Sequence Alignment/veterinary , Immunity, Innate/genetics , Base Sequence , Gene Expression Profiling/veterinary
10.
Int J Mol Sci ; 25(10)2024 May 14.
Article in English | MEDLINE | ID: mdl-38791369

ABSTRACT

Pasteurella multocida, a zoonotic pathogen that produces a 146-kDa modular toxin (PMT), causes progressive atrophic rhinitis with severe turbinate bone degradation in pigs. However, its mechanism of cytotoxicity remains unclear. In this study, we expressed PMT, purified it in a prokaryotic expression system, and found that it killed PK15 cells. The host factor CXCL8 was significantly upregulated among the differentially expressed genes in a transcriptome sequencing analysis and qPCR verification. We constructed a CXCL8-knockout cell line with a CRISPR/Cas9 system and found that CXCL8 knockout significantly increased resistance to PMT-induced cell apoptosis. CXCL8 knockout impaired the cleavage efficiency of apoptosis-related proteins, including Caspase3, Caspase8, and PARP1, as demonstrated with Western blot. In conclusion, these findings establish that CXCL8 facilitates PMT-induced PK15 cell death, which involves apoptotic pathways; this observation documents that CXCL8 plays a key role in PMT-induced PK15 cell death.


Subject(s)
Bacterial Toxins , Interleukin-8 , Pasteurella Infections , Pasteurella multocida , Animals , Apoptosis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/toxicity , Bacterial Toxins/metabolism , Caspase 8/metabolism , Caspase 8/genetics , Cell Line , CRISPR-Cas Systems , Gene Knockout Techniques , Interleukin-8/metabolism , Interleukin-8/genetics , Pasteurella multocida/genetics , Swine , Pasteurella Infections/metabolism , Pasteurella Infections/veterinary
11.
Cell Rep ; 43(5): 114240, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38753486

ABSTRACT

Adipose tissue remodeling and dysfunction, characterized by elevated inflammation and insulin resistance, play a central role in obesity-related development of type 2 diabetes (T2D) and cardiovascular diseases. Long intergenic non-coding RNAs (lincRNAs) are important regulators of cellular functions. Here, we describe the functions of linc-ADAIN (adipose anti-inflammatory), an adipose lincRNA that is downregulated in white adipose tissue of obese humans. We demonstrate that linc-ADAIN knockdown (KD) increases KLF5 and interleukin-8 (IL-8) mRNA stability and translation by interacting with IGF2BP2. Upregulation of KLF5 and IL-8, via linc-ADAIN KD, leads to an enhanced adipogenic program and adipose tissue inflammation, mirroring the obese state, in vitro and in vivo. KD of linc-ADAIN in human adipose stromal cell (ASC) hTERT adipocytes implanted into mice increases adipocyte size and macrophage infiltration compared to implanted control adipocytes, mimicking hallmark features of obesity-induced adipose tissue remodeling. linc-ADAIN is an anti-inflammatory lincRNA that limits adipose tissue expansion and lipid storage.


Subject(s)
Adipogenesis , Interleukin-8 , Kruppel-Like Transcription Factors , RNA Stability , RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Adipogenesis/genetics , Animals , RNA Stability/genetics , Interleukin-8/metabolism , Interleukin-8/genetics , Mice , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Adipocytes/metabolism , Adipose Tissue/metabolism , Obesity/metabolism , Obesity/genetics , Obesity/pathology , RNA, Messenger/metabolism , RNA, Messenger/genetics , Male , Inflammation/pathology , Inflammation/genetics , Inflammation/metabolism
12.
J Biotechnol ; 390: 1-12, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38740307

ABSTRACT

Healthcare and nutrition are facing a paradigm shift in light of advanced therapy medicinal products (ATMPs) and cellular agriculture options respectively. Both options heavily rely on some sort of animal cell culture, e.g. autologous stem cells. These cultures require various growth factors, such as interleukin-6 and 8 (IL-6/8), in a pure, safe and sustainable form that can be provided in a scalable manner. Plants seem well suited for this task because purification of small proteins can be readily achieved by membrane separation, human/animal pathogens do not replicate in plants and production can be scaled up using in-door farming or agricultural practices. Here, we illustrate this capacity by first optimizing the codon usage of IL-6/8 for translation in Nicotiana spp., as well as testing the effect of untranslated regions and product targeting to different sub-cellular compartments on expression in a high-throughput plant cell pack (PCP) assay. In the chloroplast, IL-6 accumulated up to 6.9±3.8 (SD, n=2) and 14.4±7.4 mg kg-1 (SD, n=5) were observed in case of IL-8. When transferring IL-8 expression into whole plants, accumulation was 12.3±1.5 mg kg-1 (SD, n=3). After extraction and clarification, IL-8 was purified using a two-stage process consisting of an ultrafiltration/diafiltration step with 100 kDa and 10 kDa cut off membranes followed by an IMAC polishing step. The purity, yield and recovery were 97.8%, 6.6 mg kg-1 and 38%, respectively. We evaluated the ability of the proposed purification process to remove endotoxins to ensure the compatibility of plant-made growth factors with cell culture.


Subject(s)
Interleukin-6 , Interleukin-8 , Nicotiana , Plant Cells , Interleukin-6/metabolism , Interleukin-6/genetics , Nicotiana/genetics , Nicotiana/metabolism , Plant Cells/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Plants, Genetically Modified/genetics , Humans , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
13.
Virus Res ; 345: 199391, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38754785

ABSTRACT

Coronaviruses (CoVs) are enveloped single-stranded RNA viruses that predominantly attack the human respiratory system. In recent decades, several deadly human CoVs, including SARS-CoV, SARS-CoV-2, and MERS-CoV, have brought great impact on public health and economics. However, their high infectivity and the demand for high biosafety level facilities restrict the pathogenesis research of CoV infection. Exacerbated inflammatory cell infiltration is associated with poor prognosis in CoV-associated diseases. In this study, we used human CoV 229E (HCoV-229E), a CoV associated with relatively fewer biohazards, to investigate the pathogenesis of CoV infection and the regulation of neutrophil functions by CoV-infected lung cells. Induced pluripotent stem cell (iPSC)-derived alveolar epithelial type II cells (iAECIIs) exhibiting specific biomarkers and phenotypes were employed as an experimental model for CoV infection. After infection, the detection of dsRNA, S, and N proteins validated the infection of iAECIIs with HCoV-229E. The culture medium conditioned by the infected iAECIIs promoted the migration of neutrophils as well as their adhesion to the infected iAECIIs. Cytokine array revealed the elevated secretion of cytokines associated with chemotaxis and adhesion into the conditioned media from the infected iAECIIs. The importance of IL-8 secretion and ICAM-1 expression for neutrophil migration and adhesion, respectively, was demonstrated by using neutralizing antibodies. Moreover, next-generation sequencing analysis of the transcriptome revealed the upregulation of genes associated with cytokine signaling. To summarize, we established an in vitro model of CoV infection that can be applied for the study of the immune system perturbations during severe coronaviral disease.


Subject(s)
Alveolar Epithelial Cells , Induced Pluripotent Stem Cells , Neutrophils , Humans , Neutrophils/immunology , Neutrophils/virology , Induced Pluripotent Stem Cells/virology , Alveolar Epithelial Cells/virology , COVID-19/virology , COVID-19/immunology , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , SARS-CoV-2/pathogenicity , SARS-CoV-2/physiology , SARS-CoV-2/immunology , Interleukin-8/genetics , Interleukin-8/metabolism
14.
Article in English | MEDLINE | ID: mdl-38821668

ABSTRACT

In this paper, we studied the potential genotoxic effects of human plasma from healthy volunteers, as well as patients with gastro-oesophageal reflux disease, Barrett's oesophagus (BO) and oesophageal adenocarcinoma (OAC) using the oesophageal adenocarcinoma cell line (OE33) and the lymphoblastoid cell line (TK6). Both TK6 and OE33 cells were treated with plasma (10 % volume, replacing foetal bovine serum (FBS) or horse serum (HS)) at different time points of 4 h (for the micronucleus (Mn) assay and the invasion assay) and 24 h (for the cell cycle studies). Plasma-induced effects on DNA damage levels, cell viability and the cell cycle were studied by the micronucleus assay, cytokinesis block proliferation index (CBPI) and flow cytometry respectively. The expression of IL-8 in supernatants of TK6 cells and IFN-ß in OE33 cells was also analysed by enzyme-linked immunosorbent assay (ELISA). Finally, we carried out an assessment of cellular invasion of OE33 cells following plasma treatment. The results of the micronucleus assay confirmed the genotoxicity of direct plasma treatment from some participants through the increase in DNA damage in TK6 cells. Conversely, some individual patient plasma samples reduced background levels of TK6 cell Mn frequency, in an anti-genotoxic fashion. In TK6 cells, (on average) plasma samples from patients with Barrett's oesophagus induced higher micronucleus levels than healthy volunteers (p= 0.0019). There was little difference in Mn induction when using plasma versus serum to treat the cells in vitro. Cell cycle results showed that direct plasma treatment had a marked impact on OE33 cells at 24 h (p=0.0182 for BO and p=0.0320 for OAC) by decreasing the proportion of cells in the S phase, while plasma exposure was less impactful on the cell cycle of TK6 cells. Invasion of OE33 cells was also seen to be non-significantly affected by plasma treatment of OE33 cells. The addition of N-acetyl cysteine NAC in a dose-dependent matter did not alter the formation of Mn in TK6 cells, suggesting that reactive oxygen species (ROS) are not the root cause of plasma's genotoxicity. The concentration of IL-8 in TK6 cells and IFN-ß in OE33 cells was significantly higher in cells treated with OAC-derived plasma than in the untreated negative control. Collectively, our results demonstrate that plasma-specific effects are detectable which helps us better understand some important aspects of the biology of blood-based biomarkers under development.


Subject(s)
Adenocarcinoma , Barrett Esophagus , DNA Damage , Esophageal Neoplasms , Micronucleus Tests , Humans , Barrett Esophagus/pathology , Barrett Esophagus/genetics , Adenocarcinoma/pathology , Adenocarcinoma/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Plasma/metabolism , Interleukin-8/metabolism , Interleukin-8/genetics , Cell Line, Tumor , Cell Cycle/drug effects , Male , Middle Aged , Adult , Cell Survival/drug effects , Female , Micronuclei, Chromosome-Defective , Interferon-beta , Aged
15.
Vet Res ; 55(1): 56, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38715098

ABSTRACT

The chemokine CXCL8, also known as the neutrophil chemotactic factor, plays a crucial role in mediating inflammatory responses and managing cellular immune reactions during viral infections. Porcine reproductive and respiratory syndrome virus (PRRSV) primarily infects pulmonary alveolar macrophages (PAMs), leading to acute pulmonary infections. In this study, we explored a novel long non-coding RNA (lncRNA), termed lnc-CAST, situated within the Cxcl8 gene locus. This lncRNA was found to be highly expressed in porcine macrophages. We observed that both lnc-CAST and CXCL8 were significantly upregulated in PAMs following PRRSV infection, and after treatments with lipopolysaccharide (LPS) or lipoteichoic acid (LTA). Furthermore, we noticed a concurrent upregulation of lnc-CAST and CXCL8 expression in lungs of PRRSV-infected pigs. We then determined that lnc-CAST positively influenced CXCL8 expression in PAMs. Overexpression of lnc-CAST led to an increase in CXCL8 production, which in turn enhanced the migration of epithelial cells and the recruitment of neutrophils. Conversely, inhibiting lnc-CAST expression resulted in reduced CXCL8 production in PAMs, leading to decreased migration levels of epithelial cells and neutrophils. From a mechanistic perspective, we found that lnc-CAST, localized in the nucleus, facilitated the enrichment of histone H3K27ac in CXCL8 promoter region, thereby stimulating CXCL8 transcription in a cis-regulatory manner. In conclusion, our study underscores the pivotal critical role of lnc-CAST in regulating CXCL8 production, offering valuable insights into chemokine regulation and lung damage during PRRSV infection.


Subject(s)
Histones , Interleukin-8 , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , RNA, Long Noncoding , Animals , Swine , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Porcine respiratory and reproductive syndrome virus/physiology , Interleukin-8/metabolism , Interleukin-8/genetics , Porcine Reproductive and Respiratory Syndrome/genetics , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/virology , Histones/metabolism , Histones/genetics , Macrophages, Alveolar/virology , Macrophages, Alveolar/metabolism , Gene Expression Regulation
16.
BMC Oral Health ; 24(1): 622, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38807156

ABSTRACT

BACKGROUND: C-X-C motif chemokine ligand (CXCL8), also known as interleukin-8, is a prototypical CXC family chemokine bearing a glutamic acid-leucine-arginine (ELR) motif that plays key roles in the onset and progression of a range of cancers in humans. Many prior studies have focused on exploring the relationship between CXCL8 gene polymorphisms and the risk of cancer. However, the statistical power of many of these reports was limited, yielding ambiguous or conflicting results in many cases. METHODS: Accordingly, the PubMed, Wanfang, Scopus and Web of Science databases were searched for articles published until July 20, 2023 using the keywords 'IL-8' or 'interleukin-8' or 'CXCL8', 'polymorphism' and 'cancer' or 'tumor'. Odds ratios (ORs) and 95% confidence intervals (CIs) were utilized to examine the association. The CXCL8 +781 polymorphism genotypes were assessed with a TaqMan assay. RESULTS: About 29 related publications was conducted in an effort to better understand the association between these polymorphisms and disease risk. The CXCL8 -353A/T polymorphism was associated with an increased overall cancer risk [A vs. T, odds ratio (OR) = 1.255, 95% confidence interval (CI) (1.079-1.459), Pheterogeneity = 0.449, P = 0.003]. The CXCL8 +781 T/C allele was similarly associated with a higher risk of cancer among Caucasians [TT vs. TC + CC, OR = 1.320, 95%CI (1.046-1.666), Pheterogeneity = 0.375, P = 0.019]. Furthermore, oral cancer patients carrying the CXCL8 +781 TT + TC genotypes exhibited pronounced increases in serum levels of CXCL8 as compared to the CC genotype (P < 0.01), and also shown similar trend as compared to genotype-matched normal controls (P < 0.01). Finally, several limitations, such as the potential for publication bias or heterogeneity among the included studies should be paid attention. CONCLUSION: Current study suggested that the CXCL8 -353 and +781 polymorphisms may be associated with a greater risk of cancer, which might impact cancer prevention, diagnosis, or treatment through the different expression of CXCL8. At the same time, the +781 polymorphism may further offer value as a biomarker that can aid in the early identification and prognostic evaluation of oral cancer.


Subject(s)
Genetic Predisposition to Disease , Interleukin-8 , Mouth Neoplasms , Humans , Interleukin-8/genetics , Mouth Neoplasms/genetics , Case-Control Studies , Genetic Predisposition to Disease/genetics , Polymorphism, Genetic , Polymorphism, Single Nucleotide , Risk Factors
17.
Environ Toxicol Pharmacol ; 108: 104469, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38759848

ABSTRACT

We analyzed gene expression in THP-1 cells exposed to metal-based nanomaterials (NMs) [TiO2 (NM-100), ZnO (NM-110), SiO2 (NM-200), Ag (NM-300 K)]. A functional enrichment analysis of the significant differentially expressed genes (DEGs) identified the key modulated biological processes and pathways. DEGs were used to construct protein-protein interaction networks. NM-110 and NM-300 K induced changes in the expression of genes involved in oxidative and genotoxic stress, immune response, alterations of cell cycle, detoxification of metal ions and regulation of redox-sensitive pathways. Both NMs shared a number of highly connected protein nodes (hubs) including CXCL8, ATF3, HMOX1, and IL1B. NM-200 induced limited transcriptional changes, mostly related to the immune response; however, several hubs (CXCL8, ATF3) were identical with NM-110 and NM-300 K. No effects of NM-100 were observed. Overall, soluble nanomaterials NM-110 and NM-300 K exerted a wide variety of toxic effects, while insoluble NM-200 induced immunotoxicity; NM-100 caused no detectable changes on the gene expression level.


Subject(s)
Protein Interaction Maps , Silver , Titanium , Humans , Titanium/toxicity , THP-1 Cells , Protein Interaction Maps/drug effects , Silver/toxicity , Nanostructures/toxicity , Metal Nanoparticles/toxicity , Zinc Oxide/toxicity , Zinc Oxide/chemistry , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Transcriptome/drug effects , Silicon Dioxide/toxicity , Interleukin-8/metabolism , Interleukin-8/genetics , Heme Oxygenase-1
18.
J Clin Invest ; 134(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38747289

ABSTRACT

CXCL8 and other chemokines have been implicated in tissue inflammation and are attractive candidates for therapeutic targeting to treat human disease.


Subject(s)
Interleukin-8 , Humans , Interleukin-8/metabolism , Interleukin-8/genetics , Animals , Inflammation/immunology , Inflammation/metabolism
19.
Cancer Lett ; 591: 216893, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38636892

ABSTRACT

The oncogenic properties of Nucleobindin2 (NUCB2) have been observed in various cancer types. Nevertheless, the precise understanding of the biological functions and regulatory mechanisms of NUCB2 in osteosarcoma remains limited. This investigation reported that NUCB2 was significantly increased upon glucose deprivation-induced metabolic stress. Elevated NUCB2 suppressed glucose deprivation-induced cell death and reactive oxygen species (ROS) increase. Depletion of NUCB2 resulted in a reduction in osteosarcoma cell proliferation as well as metastatic potential in vitro and in vivo. Mechanically, NUCB2 ablation suppressed C-X-C Motif Chemokine Ligand 8 (CXCL8) expression which then reduced programmed cell death 1 ligand 1 (PD-L1) expression and stimulated anti-tumor immunity mediated through cytotoxic T cells. Importantly, a combination of NUCB2 depletion with anti-PD-L1 treatment improved anti-tumor T-cell immunity in vivo. Moreover, we further demonstrated that NUCB2 interacted with NUCKS1 to inhibit its degradation, which is responsible for the transcriptional regulation of CXCL8 expression. Altogether, the outcome emphasizes the function of NUCB2 in osteosarcoma and indicates that NUCB2 elevates osteosarcoma progression and immunosuppressive microenvironment through the NUCKS1/CXCL8 pathway.


Subject(s)
Bone Neoplasms , Calcium-Binding Proteins , Disease Progression , Interleukin-8 , Osteosarcoma , Tumor Microenvironment , Osteosarcoma/immunology , Osteosarcoma/pathology , Osteosarcoma/metabolism , Osteosarcoma/genetics , Humans , Animals , Cell Line, Tumor , Interleukin-8/metabolism , Interleukin-8/genetics , Mice , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Bone Neoplasms/genetics , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Tumor Microenvironment/immunology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Cell Proliferation , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Gene Expression Regulation, Neoplastic , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , T-Lymphocytes, Cytotoxic/immunology , Signal Transduction , Reactive Oxygen Species/metabolism
20.
Int Immunopharmacol ; 133: 112065, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38608448

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) functions to regulate inflammation and immune response, but its mechanism is not fully understood. We report here that STAT3 inhibitors Stattic and Niclosamide up-regulated IL-1ß-induced IL-8 production in C33A, CaSki, and Siha cervical cancer cells. As expected, IL-1ß-induced IL-8 production was also up-regulated through the molecular inhibition of STAT3 by use of CRISPR/Cas9 technology. Unexpectedly, IL-1ß induced IL-8 production via activating ERK and P38 signal pathways, but neither STAT3 inhibitors nor STAT3 knockout affected IL-1ß-induced signal transduction, suggesting that STAT3 decreases IL-8 production not via inhibition of signal transduction. To our surprise, STAT3 inhibition increased the stabilization, and decreased the degradation of IL-8 mRNA, suggesting a post-transcriptional regulation of IL-1ß-induced IL-8. Moreover, Dihydrotanshinone I, an inhibitor of RNA-binding protein HuR, down-regulated IL-1ß-induced IL-8 dose-dependently. HuR inhibition by CRISPR/Cas9 also decreased IL-8 production induced by IL-1ß. Mechanistically, co-immunoprecipitation results showed that STAT3 did not react with HuR directly, but STAT3 inhibition increased the protein levels of HuR in cytoplasm. And IL-6 activation of STAT3 induced HuR cytoplasmic-nuclear transport. Taken together, these results suggest that STAT3 contributes to HuR nuclear localization and inhibits Il-1ß-induced IL-8 production through this non-transcriptional mechanism.


Subject(s)
Cell Nucleus , Cytoplasm , ELAV-Like Protein 1 , Interleukin-1beta , Interleukin-8 , STAT3 Transcription Factor , Humans , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Interleukin-1beta/metabolism , Interleukin-8/metabolism , Interleukin-8/genetics , ELAV-Like Protein 1/metabolism , ELAV-Like Protein 1/genetics , Cytoplasm/metabolism , Cell Nucleus/metabolism , Cell Line, Tumor , Cyclic S-Oxides/pharmacology , Protein Transport , Signal Transduction , Active Transport, Cell Nucleus , CRISPR-Cas Systems
SELECTION OF CITATIONS
SEARCH DETAIL