Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 23.121
Filtrer
1.
Clin Epigenetics ; 16(1): 136, 2024 Oct 02.
Article de Anglais | MEDLINE | ID: mdl-39358792

RÉSUMÉ

Osteosarcoma (OS) is the most frequent primary malignant bone tumour, whose heterogeneity represents a major challenge for common antitumour therapies. Inflammatory cytokines are known to be necessary for OS progression. Therefore, to optimise therapy, it is important to discover reliable biomarkers by identifying the mechanism generating OS and investigating the inflammatory pathways that support the undifferentiated state. In this work, we highlight the differences of epigenetic activities of IL-1ß and TNFα, and the susceptibility of TET-1 enzymatic inhibition, in tumour progression of three different OS cell lines. Investigating DNA methylation of IL-6 promoter and determining its expression, we found that TET enzymatic inhibition influences proliferation induced by inflammatory cytokines in OS cell lines. Moreover, Bobcat 339 treatment blocks IL-1ß epigenetic action on IL-6 promoter, while only partially those of TNFα as well as inhibits IL-1ß-dependent epithelial-mesenchymal transition (EMT) process, but only partially those of TNFα. In conclusion, this work highlights that IL-1ß and TNFα have different effects on DNA demethylation in OS cell lines, making DNA methylation a potential biomarker of disease. Specifically, in IL-1ß treatment, TET-1 inhibition completely blocks tumour progression, while in TNFα actions, it is only partially effective. Given that these two inflammatory pathways can be therapeutic targets for treating these tumours, knowledge of their distinct epigenetic behaviours can be useful for developing precise and specific therapeutic strategies for this disease.


Sujet(s)
Méthylation de l'ADN , Épigenèse génétique , Interleukine-1 bêta , Ostéosarcome , Protéines proto-oncogènes , Facteur de nécrose tumorale alpha , Humains , Interleukine-1 bêta/génétique , Interleukine-1 bêta/pharmacologie , Épigenèse génétique/effets des médicaments et des substances chimiques , Épigenèse génétique/génétique , Facteur de nécrose tumorale alpha/génétique , Facteur de nécrose tumorale alpha/pharmacologie , Méthylation de l'ADN/génétique , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Protéines proto-oncogènes/génétique , Ostéosarcome/génétique , Ostéosarcome/traitement médicamenteux , Évolution de la maladie , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Mixed function oxygenases/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/génétique , Interleukine-6/génétique , Tumeurs osseuses/génétique , Tumeurs osseuses/traitement médicamenteux , Tumeurs osseuses/anatomopathologie
2.
Int J Mol Sci ; 25(17)2024 Aug 23.
Article de Anglais | MEDLINE | ID: mdl-39273085

RÉSUMÉ

Inflammation models are widely used in the in vitro investigation of new therapeutic approaches for osteoarthritis. TNFα (tumor necrosis factor alpha) plays an important role in the inflammatory process. Current inflammation models lack uniformity and make comparisons difficult. Therefore, this study aimed to systematically investigate whether the effects of TNFα are concentration-dependent and whether chondrocyte expansion has an effect on the inflammatory model. Bovine chondrocytes were enzymatically isolated, expanded to passages 1-3, and transferred into a 3D pellet culture. Chondrocyte pellets were stimulated with recombinant bovine TNFα at different concentrations for 48 h to induce inflammation. Gene expression of anabolic (collagen 2, aggrecan, cartilage oligomeric protein (COMP)), catabolic (matrix metalloproteinases (MMP3, MMP13)), dedifferentiation (collagen 1) markers, inflammation markers (interleukin-6 (IL-6), nuclear factor kappa B (NFkB), cyclooxygenase-2 (COX), prostaglandin-E-synthase-2 (PTGES2)), and the apoptosis marker caspase 3 was determined. At the protein level, concentrations of IL-6, nitric oxide (NO), and sulfated glycosaminoglycans (GAG) were evaluated. Statistical analysis was performed using the independent t-test, and significance was defined as p < 0.05. In general, TNFα caused a decrease in anabolic markers and an increase in the expression of catabolic and inflammatory markers. There was a concentration-dependent threshold of 10 ng/mL to induce significant inflammatory effects. Most of the markers analyzed showed TNFα concentration-dependent effects (COMP, PRG4, AGN, Col1, MMP3, and NFkB). There was a statistical influence of selected gene expression markers from different passages on the TNFα chondrocyte inflammation model, including Col2, MMP13, IL-6, NFkB, COX2, and PTGES2. Considering the expression of collagen 2 and MMP3, passage 3 chondrocytes showed a higher sensitivity to TNFα stimulation compared to passages 1 and 2. On the other hand, MMP13, IL-6, NFkB, and caspase 3 gene expression were lower in P3 chondrocytes compared to the other passages. On the protein level, inflammatory effects showed a similar pattern, with cytokine effects starting at 10 ng/mL and differences between the passages. TNFα had a detrimental effect on cartilage, with a clear threshold observed at 10 ng/mL. Although TNFα effects showed concentration-dependent patterns, this was not consistent for all markers. The selected passage showed a clear influence, especially on inflammation markers. Further experiments were warranted to explore the effects of TNFα concentration and passage in long-term stimulation.


Sujet(s)
Chondrocytes , Inflammation , Facteur de nécrose tumorale alpha , Animaux , Chondrocytes/métabolisme , Chondrocytes/effets des médicaments et des substances chimiques , Bovins , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Inflammation/métabolisme , Inflammation/anatomopathologie , Cellules cultivées , Facteur de transcription NF-kappa B/métabolisme , Monoxyde d'azote/métabolisme , Interleukine-6/métabolisme , Interleukine-6/génétique , Marqueurs biologiques
3.
Physiol Rep ; 12(18): e70063, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39327065

RÉSUMÉ

The transplantation of collagen hydrogels encapsulating human dental pulp stem cell (DPSC)-derived chondrogenic cells is potentially a novel approach for the regeneration of degenerated nucleus pulposus (NP) and cartilage. Grafted cell migration allows cells to disperse in the hydrogels and the treated tissue from the grafted location. We previously reported the cell migration in type I and type II hydrogels. It is important to explore further how cell culture time affect the cell motility. In this study, we observed the decreased motility of DPSC-derived chondrogenic cells after culturing for 2 weeks compared with cells cultured for 2 days in these gels. The Alamarblue assay showed the cell proliferation during the two-week cell culture period. The findings suggest that the transitions of cell motility and proliferation during the longer culture time. The result indicates that the early culture stage is an optimal time for cell transplantation. In a degenerated disc, the expression of IL-1ß and TNFα increased significantly compared with healthy tissue and therefore may affect grafted cell migration. The incorporation of IL-1ß and TNFα into the collagen hydrogels decreased cell motility. The study indicates that the control of IL-1ß and TNFα production may help to maintain cell motility after transplantation.


Sujet(s)
Mouvement cellulaire , Collagène , Pulpe dentaire , Hydrogels , Cellules souches , Pulpe dentaire/cytologie , Humains , Cellules souches/cytologie , Cellules souches/métabolisme , Collagène/métabolisme , Cellules cultivées , Prolifération cellulaire , Chondrogenèse , Interleukine-1 bêta/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Adulte , Adolescent , Cytokines/métabolisme , Techniques de culture cellulaire/méthodes , Jeune adulte
4.
Biochim Biophys Acta Mol Basis Dis ; 1870(8): 167479, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39181516

RÉSUMÉ

Intracellular cholesterol metabolism is regulated by the SREBP-2 and LXR signaling pathways. The effects of inflammation on these molecular mechanisms remain poorly studied, especially at the blood-brain barrier (BBB) level. Tumor necrosis factor α (TNFα) is a proinflammatory cytokine associated with BBB dysfunction. Therefore, the aim of our study was to investigate the effects of TNFα on BBB cholesterol metabolism, focusing on its underlying signaling pathways. Using a human in vitro BBB model composed of human brain-like endothelial cells (hBLECs) and brain pericytes (HBPs), we observed that TNFα increases BBB permeability by degrading the tight junction protein CLAUDIN-5 and activating stress signaling pathways in both cell types. TNFα also promotes cholesterol release and decreases cholesterol accumulation and APOE secretion. In hBLECs, the expression of SREBP-2 targets (LDLR and HMGCR) is increased, while ABCA1 expression is decreased. In HBPs, only LDLR and ABCA1 expression is increased. TNFα treatment also induces 25-hydroxycholesterol (25-HC) production, a cholesterol metabolite involved in the immune response and intracellular cholesterol metabolism. 25-HC pretreatment attenuates TNFα-induced BBB leakage and partially alleviates the effects of TNFα on ABCA1, LDLR, and HMGCR expression. Overall, our results suggest that TNFα favors cholesterol efflux via an LXR/ABCA1-independent mechanism at the BBB, while it activates the SREBP-2 pathway. Treatment with 25-HC partially reversed the effect of TNFα on the LXR/SREBP-2 pathways. Our study provides novel perspectives for better understanding cerebrovascular signaling events linked to BBB dysfunction and cholesterol metabolism in neuroinflammatory diseases.


Sujet(s)
Barrière hémato-encéphalique , Cholestérol , Cellules endothéliales , Hydroxycholestérols , Protéine-2 de liaison à l'élément de régulation des stérols , Facteur de nécrose tumorale alpha , Hydroxycholestérols/pharmacologie , Hydroxycholestérols/métabolisme , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Humains , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Protéine-2 de liaison à l'élément de régulation des stérols/métabolisme , Protéine-2 de liaison à l'élément de régulation des stérols/génétique , Cellules endothéliales/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Cholestérol/métabolisme , Récepteurs aux lipoprotéines LDL/métabolisme , Récepteurs aux lipoprotéines LDL/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Membre-1 de la sous-famille A des transporteurs à cassette liant l'ATP/métabolisme , Membre-1 de la sous-famille A des transporteurs à cassette liant l'ATP/génétique , Péricytes/métabolisme , Péricytes/effets des médicaments et des substances chimiques , Péricytes/anatomopathologie , Hydroxymethylglutaryl-CoA reductases/métabolisme , Hydroxymethylglutaryl-CoA reductases/génétique , Apolipoprotéines E/métabolisme , Apolipoprotéines E/génétique , Récepteurs hépatiques X/métabolisme , Récepteurs hépatiques X/génétique , Cellules cultivées
5.
Int J Mol Sci ; 25(16)2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39201392

RÉSUMÉ

Laminins are essential components of the basement membranes, expressed in a tissue- and cell-specific manner under physiological conditions. During inflammatory circumstances, such as atherosclerosis, alterations in laminin composition within vessels have been observed. Our study aimed to assess the influence of tumor necrosis factor-alpha (TNF), a proinflammatory cytokine abundantly found in atherosclerotic lesions, on endothelial laminin gene expression and the effects of laminin-332 (LN332) on endothelial cells' behavior. We also evaluated the expression of LN332-encoding genes in human carotid atherosclerotic plaques. Our findings demonstrate that TNF induces upregulation of LAMB3 and LAMC2, which, along with LAMA3, encode the LN332 isoform. Endothelial cells cultured on recombinant LN332 exhibit decreased claudin-5 expression and display a loosely connected phenotype, with an elevated expression of chemokines and leukocyte adhesion molecules, enhancing their attractiveness and adhesion to leukocytes in vitro. Furthermore, LAMB3 and LAMC2 are upregulated in human carotid plaques and show a positive correlation with TNF expression. In summary, TNF stimulates the expression of LN332-encoding genes in human endothelial cells and LN332 promotes an endothelial phenotype characterized by compromised junctional integrity and increased leukocyte interaction. These findings highlight the importance of basement membrane proteins for endothelial integrity and the potential role of LN332 in atherosclerosis.


Sujet(s)
Athérosclérose , Molécules d'adhérence cellulaire , , Laminine , Facteur de nécrose tumorale alpha , Humains , Facteur de nécrose tumorale alpha/pharmacologie , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/génétique , Molécules d'adhérence cellulaire/génétique , Molécules d'adhérence cellulaire/métabolisme , Athérosclérose/génétique , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Laminine/métabolisme , Laminine/génétique , Cellules endothéliales/métabolisme , Phénotype , Plaque d'athérosclérose/génétique , Plaque d'athérosclérose/métabolisme , Plaque d'athérosclérose/anatomopathologie , Adhérence cellulaire/génétique , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Cellules cultivées
6.
Cell Death Dis ; 15(8): 584, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39122678

RÉSUMÉ

TNF is a potent cytokine known for its involvement in physiology and pathology. In Rheumatoid Arthritis (RA), persistent TNF signals cause aberrant activation of synovial fibroblasts (SFs), the resident cells crucially involved in the inflammatory and destructive responses of the affected synovial membrane. However, the molecular switches that control the pathogenic activation of SFs remain poorly defined. Cyld is a major component of deubiquitination (DUB) machinery regulating the signaling responses towards survival/inflammation and programmed necrosis that induced by cytokines, growth factors and microbial products. Herein, we follow functional genetic approaches to understand how Cyld affects arthritogenic TNF signaling in SFs. We demonstrate that in spontaneous and induced RA models, SF-Cyld DUB deficiency deteriorates arthritic phenotypes due to increased levels of chemokines, adhesion receptors and bone-degrading enzymes generated by mutant SFs. Mechanistically, Cyld serves to restrict the TNF-induced hyperactivation of SFs by limiting Tak1-mediated signaling, and, therefore, leading to supervised NF-κB and JNK activity. However, Cyld is not critically involved in the regulation of TNF-induced death of SFs. Our results identify SF-Cyld as a regulator of TNF-mediated arthritis and inform the signaling landscape underpinning the SF responses.


Sujet(s)
Polyarthrite rhumatoïde , Deubiquitinating enzyme CYLD , Fibroblastes , I-kappa B Kinase , MAP Kinase Kinase Kinases , Transduction du signal , Membrane synoviale , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Deubiquitinating enzyme CYLD/métabolisme , Deubiquitinating enzyme CYLD/génétique , MAP Kinase Kinase Kinases/métabolisme , MAP Kinase Kinase Kinases/génétique , Animaux , Membrane synoviale/métabolisme , Membrane synoviale/anatomopathologie , Souris , I-kappa B Kinase/métabolisme , I-kappa B Kinase/génétique , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/génétique , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Humains , Facteur de transcription NF-kappa B/métabolisme , Souris de lignée C57BL , Souris knockout
7.
Int J Mol Sci ; 25(15)2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-39126115

RÉSUMÉ

Connexin 43 (Cx43) is crucial for the development and homeostasis of the musculoskeletal system, where it plays multifaceted roles, including intercellular communication, transcriptional regulation and influencing osteogenesis and chondrogenesis. Here, we investigated Cx43 modulation mediated by inflammatory stimuli involved in osteoarthritis, i.e., 10 ng/mL Tumor Necrosis Factor alpha (TNFα) and/or 1 ng/mL Interleukin-1 beta (IL-1ß), in primary chondrocytes (CH) and osteoblasts (OB). Additionally, we explored the impact of synovial fluids from osteoarthritis patients in CH and cartilage explants, providing a more physio-pathological context. The effect of TNFα on Cx43 expression in cartilage explants was also assessed. TNFα downregulated Cx43 levels both in CH and OB (-73% and -32%, respectively), while IL-1ß showed inconclusive effects. The reduction in Cx43 levels was associated with a significant downregulation of the coding gene GJA1 expression in OB only (-65%). The engagement of proteasome in TNFα-induced effects, already known in CH, was also observed in OB. TNFα treatment significantly decreased Cx43 expression also in cartilage explants. Of note, Cx43 expression was halved by synovial fluid in both CH and cartilage explants. This study unveils the regulation of Cx43 in diverse musculoskeletal cell types under various stimuli and in different contexts, providing insights into its modulation in inflammatory joint disorders.


Sujet(s)
Chondrocytes , Connexine 43 , Interleukine-1 bêta , Arthrose , Ostéoblastes , Facteur de nécrose tumorale alpha , Humains , Connexine 43/métabolisme , Connexine 43/génétique , Chondrocytes/métabolisme , Ostéoblastes/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/pharmacologie , Arthrose/métabolisme , Arthrose/anatomopathologie , Arthrose/génétique , Synovie/métabolisme , Cartilage articulaire/métabolisme , Cartilage articulaire/anatomopathologie , Cellules cultivées , Sujet âgé , Adulte d'âge moyen , Inflammation/métabolisme , Inflammation/génétique , Inflammation/anatomopathologie , Cartilage/métabolisme , Cartilage/anatomopathologie , Maladies articulaires/métabolisme , Maladies articulaires/anatomopathologie , Maladies articulaires/génétique
8.
Cells ; 13(13)2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38995013

RÉSUMÉ

Skeletal muscle regeneration after injury is a complex process involving inflammatory signaling and myoblast activation. Pro-inflammatory cytokines like tumor necrosis factor-alpha (TNF-α) are key mediators, but their effects on gene expression in proliferating myoblasts are unclear. We performed the RNA sequencing of TNF-α treated C2C12 myoblasts to elucidate the signaling pathways and gene networks regulated by TNF-α during myoblast proliferation. The TNF-α (10 ng/mL) treatment of C2C12 cells led to 958 differentially expressed genes compared to the controls. Pathway analysis revealed significant regulation of TNF-α signaling, along with the chemokine and IL-17 pathways. Key upregulated genes included cytokines (e.g., IL-6), chemokines (e.g., CCL7), and matrix metalloproteinases (MMPs). TNF-α increased myogenic factor 5 (Myf5) but decreased MyoD protein levels and stimulated the release of MMP-9, MMP-10, and MMP-13. TNF-α also upregulates versican and myostatin mRNA. Overall, our study demonstrates the TNF-α modulation of distinct gene expression patterns and signaling pathways that likely contribute to enhanced myoblast proliferation while suppressing premature differentiation after muscle injury. Elucidating the mechanisms involved in skeletal muscle regeneration can aid in the development of regeneration-enhancing therapeutics.


Sujet(s)
Prolifération cellulaire , Myoblastes , Transduction du signal , Facteur de nécrose tumorale alpha , Myoblastes/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Animaux , Souris , Lignée cellulaire , Chimiokines/métabolisme , Chimiokines/génétique , Cytokines/métabolisme , Cytokines/génétique , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques
9.
Front Immunol ; 15: 1435701, 2024.
Article de Anglais | MEDLINE | ID: mdl-39044828

RÉSUMÉ

Ceramides generated by the activity of the neutral sphingomyelinase 2 (nSMase2) play a pivotal role in stress responses in mammalian cells. Dysregulation of sphingolipid metabolism has been implicated in numerous inflammation-related pathologies. However, its influence on inflammatory cytokine-induced signaling is yet incompletely understood. Here, we used proximity labeling to explore the plasma membrane proximal protein network of nSMase2 and TNFα-induced changes thereof. We established Jurkat cells stably expressing nSMase2 C-terminally fused to the engineered ascorbate peroxidase 2 (APEX2). Removal of excess biotin phenol substantially improved streptavidin-based affinity purification of biotinylated proteins. Using our optimized protocol, we determined nSMase2-proximal biotinylated proteins and their changes within the first 5 min of TNFα stimulation by quantitative mass spectrometry. We observed significant dynamic changes in the nSMase2 microenvironment in response to TNFα stimulation consistent with rapid remodeling of protein networks. Our data confirmed known nSMase2 interactors and revealed that the recruitment of most proteins depended on nSMase2 enzymatic activity. We measured significant enrichment of proteins related to vesicle-mediated transport, including proteins of recycling endosomes, trans-Golgi network, and exocytic vesicles in the proximitome of enzymatically active nSMase2 within the first minutes of TNFα stimulation. Hence, the nSMase2 proximal network and its TNFα-induced changes provide a valuable resource for further investigations into the involvement of nSMase2 in the early signaling pathways triggered by TNFα.


Sujet(s)
Sphingomyeline phosphodiesterase , Facteur de nécrose tumorale alpha , Humains , Facteur de nécrose tumorale alpha/pharmacologie , Facteur de nécrose tumorale alpha/métabolisme , Cellules Jurkat , Sphingomyeline phosphodiesterase/métabolisme , Transduction du signal , Membrane cellulaire/métabolisme
10.
Int J Mol Sci ; 25(13)2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-39000465

RÉSUMÉ

The complexification of in vitro models requires the compatibility of cells with the same medium. Since immune cells are the most sensitive to growth conditions, growing intestinal epithelial cells in their usual medium seems to be necessary. This work was aimed at comparing the sensitivity of these epithelial cells to pro-inflammatory stimuli but also to dietary polyphenols in both DMEM and RPMI-1640 media. Co-cultures of Caco-2 and HT29-MTX cells were grown for 21 days in the two media before their stimulation with a cocktail of TNF-α (20 ng/mL), IL-1ß (1 ng/mL), and IFN-γ (10 ng/mL) or with LPS (10 ng/mL) from E. coli (O111:B4). The role of catechins (15 µM), a dietary polyphenol, was evaluated after its incubation with the cells before their stimulation for 6 h. The RPMI-1640 medium did not alter the intensity of the inflammatory response observed with the cytokines. By contrast, LPS failed to stimulate the co-culture in inserts regardless of the medium used. Lastly, catechins were unable to prevent the pro-inflammatory response observed with the cytokines in the two media. The preservation of the response of this model of intestinal epithelium in RPMI-1640 medium is promising when considering its complexification to evaluate the complex cellular crosstalk leading to intestinal homeostasis.


Sujet(s)
Techniques de coculture , Muqueuse intestinale , Lipopolysaccharides , Polyphénols , Humains , Techniques de coculture/méthodes , Polyphénols/pharmacologie , Cellules Caco-2 , Muqueuse intestinale/métabolisme , Muqueuse intestinale/cytologie , Muqueuse intestinale/effets des médicaments et des substances chimiques , Lipopolysaccharides/pharmacologie , Cellules HT29 , Milieux de culture/composition chimique , Milieux de culture/pharmacologie , Cytokines/métabolisme , Catéchine/pharmacologie , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Inflammation/métabolisme , Inflammation/anatomopathologie
11.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167355, 2024 10.
Article de Anglais | MEDLINE | ID: mdl-39009172

RÉSUMÉ

BACKGROUND: HOIP is the catalytic subunit of the E3 ligase complex (linear ubiquitin chain assembly complex), which is able to generate linear ubiquitin chains. However, the role of rare HOIP functionally deficient variants remains unclear. The pathogenic mechanism and the relationship with immune deficiency phenotypes remain to be clarified. METHODS: Based on a next-generation sequencing panel of 270 genes, we identified a HOIP deletion variant that causes common variable immunodeficiency disease. Bioinformatics analysis and cell-based experiments were performed to study the molecular mechanism by which the variant causes immunodeficiency diseases. FINDINGS: A homozygous loss-of-function variant in HOIP was identified. The variant causes a frameshift and generates a premature termination codon in messenger RNA, resulting in a C-terminal truncated HOIP mutant, that is, the loss of the linear ubiquitin chain-specific catalytic domain. The truncated HOIP mutant has impaired E3 ligase function in linear ubiquitination, leading to the suppression of canonical NF-κB signalling and increased TNF-induced multiple forms of cell death. INTERPRETATION: The loss-of-function HOIP variant accounts for the immune deficiencies. The canonical NF-κB pathway and cell death are involved in the pathogenesis of the disease. FUNDING: This study was funded by the National Natural Science Foundation of China (No. 82270444 and 81501851). RESEARCH IN CONTEXT: Evidence before this study LUBAC is the only known linear ubiquitin chain assembly complex for which HOIP is an essential catalytic subunit. Three HOIP variants have now been identified in two immunodeficient patients and functionally characterised. However, there have been no reports on the pathogenicity of only catalytic domain deletion variants in humans, or the pathogenic mechanisms of catalytic domain deletion variants. Added value of this study We report the first case of an autosomal recessive homozygous deletion variant that results in deletion of the HOIP catalytic structural domain. We demonstrate that this variant is a loss-of-function variant using a heterologous expression system. The variant has impaired E3 ligase function. It can still bind to other subunits of LUBAC, but it fails to generate linear ubiquitin chains. We also explored the underlying mechanisms by which this variant leads to immunodeficiency. The variant attenuates the canonical NF-κB and MAPK signalling cascades and increases the sensitivity of TNFα-induced diverse cell death and activation of mitochondrial apoptosis pathways. These findings provide support for the treatment and drug development of patients with inborn errors of immunity in HOIP and related signalling pathways. Implications of all the available evidence First, this study expands the HOIP pathogenic variant database and phenotypic spectrum. Furthermore, studies on the biological functions of pathogenic variants in relation to the NF-κB signalling pathway and cell death provided new understanding into the genetic basis and pathogenesis of HOIP-deficient immune disease, indicating the necessity of HOIP and related signalling pathway variants as diagnostic targets in patients with similar genetic deficiency phenotypes..


Sujet(s)
Mutation avec décalage du cadre de lecture , Facteur de transcription NF-kappa B , Transduction du signal , Facteur de nécrose tumorale alpha , Ubiquitin-protein ligases , Femelle , Humains , Mâle , Mort cellulaire/effets des médicaments et des substances chimiques , Mort cellulaire/génétique , Cellules HEK293 , Facteur de transcription NF-kappa B/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Ubiquitination , Enfant , Pedigree
12.
Biochem Biophys Res Commun ; 727: 150316, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-38959732

RÉSUMÉ

Type 2 diabetes (T2D) is on a notable rise worldwide, which leads to unfavorable outcomes during implant treatments. Surface modification of implants and exosome treatment have been utilized to enhance osseointegration. However, there has been insufficient approach to improve adverse osseointegration in T2D conditions. In this study, we successfully loaded TNF-α-treated mesenchymal stem cell (MSC)-derived exosomes onto micro/nano-network titanium (Ti) surfaces. TNF-α-licensed exosome-integrated titanium (TNF-exo-Ti) effectively enhanced M2 macrophage polarization in hyperglycemic conditions, with increased secretion of anti-inflammatory cytokines and decreased secretion of pro-inflammatory cytokines. In addition, TNF-exo-Ti pretreated macrophage further enhanced angiogenesis and osteogenesis of endothelial cells and bone marrow MSCs. More importantly, TNF-exo-Ti markedly promoted osseointegration in T2D mice. Mechanistically, TNF-exo-Ti activated macrophage autophagy to promote M2 polarization through inhibition of the PI3K/AKT/mTOR pathway, which could be abolished by PI3K agonist. Thus, this study established TNF-α-licensed exosome-immobilized titanium surfaces that could rectify macrophage immune states and accelerate osseointegration in T2D conditions.


Sujet(s)
Autophagie , Diabète de type 2 , Exosomes , Macrophages , Souris de lignée C57BL , Ostéo-intégration , Titane , Facteur de nécrose tumorale alpha , Titane/composition chimique , Titane/pharmacologie , Animaux , Exosomes/métabolisme , Autophagie/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Ostéo-intégration/effets des médicaments et des substances chimiques , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Souris , Diabète de type 2/métabolisme , Mâle , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/cytologie , Polarité de la cellule/effets des médicaments et des substances chimiques , Ostéogenèse/effets des médicaments et des substances chimiques , Activation des macrophages/effets des médicaments et des substances chimiques , Diabète expérimental/métabolisme
13.
Stem Cell Reports ; 19(8): 1092-1106, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39059376

RÉSUMÉ

Microglia (µG), the resident immune cells in the central nervous system, surveil the parenchyma to maintain the structural and functional homeostasis of neurons. Besides, they influence neurogenesis and synaptogenesis through complement-mediated phagocytosis. Emerging evidence suggests that µG may also influence development through proinflammatory cytokines. Here, we examined the premise that tumor necrosis factor alpha (TNF-α) and interleukin-1ß (IL-1ß), the two most prominent components of the µG secretome, influence retinal development, specifically the morphological and functional differentiation of human retinal ganglion cells (hRGCs). Using controlled generation of hRGCs and human µG (hµG) from pluripotent stem cells, we demonstrate that TNF-α and IL-1ß secreted by unchallenged hµG did not influence hRGC generation. However, their presence significantly facilitated neuritogenesis along with the basal function of hRGCs, which involved the recruitment of the AKT/mTOR pathway. We present ex vivo evidence that proinflammatory cytokines may play an important role in the morphological and physiological maturation of hRGCs, which may be recapitulated for regeneration.


Sujet(s)
Différenciation cellulaire , Cytokines , Microglie , Cellules ganglionnaires rétiniennes , Humains , Cellules ganglionnaires rétiniennes/métabolisme , Cellules ganglionnaires rétiniennes/cytologie , Microglie/métabolisme , Microglie/cytologie , Cytokines/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Protéines proto-oncogènes c-akt/métabolisme , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/pharmacologie , Transduction du signal , Sérine-thréonine kinases TOR/métabolisme , Neurogenèse , Régénération , Médiateurs de l'inflammation/métabolisme , Régénération nerveuse
14.
Sci Rep ; 14(1): 16032, 2024 07 11.
Article de Anglais | MEDLINE | ID: mdl-38992075

RÉSUMÉ

This study explores the application of the RIP3-caspase3-assay in heterogeneous spheroid cultures to analyze cell death pathways, emphasizing the nuanced roles of apoptosis and necroptosis. By employing directly conjugated monoclonal antibodies, we provide detailed insights into the complex mechanisms of cell death. Our findings demonstrate the assay's capability to differentiate between RIP1-independent apoptosis, necroptosis, and RIP1-dependent apoptosis, marking a significant advancement in organoid research. Additionally, we investigate the effects of TNFα on isolated intestinal epithelial cells, revealing a concentration-dependent response and an adaptive or threshold reaction to TNFα-induced stress. The results indicate a preference for RIP1-independent cell death pathways upon TNFα stimulation, with a notable increase in apoptosis and a secondary role of necroptosis. Our research underscores the importance of the RIP3-caspase3-assay in understanding cell death mechanisms in organoid cultures, offering valuable insights for disease modeling and the development of targeted therapies. The assay's adaptability and robustness in spheroid cultures enhances its potential as a tool in personalized medicine and translational research.


Sujet(s)
Apoptose , Caspase-3 , Nécroptose , Receptor-Interacting Protein Serine-Threonine Kinases , Sphéroïdes de cellules , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Humains , Sphéroïdes de cellules/métabolisme , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Caspase-3/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Nécroptose/effets des médicaments et des substances chimiques , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Mort cellulaire/effets des médicaments et des substances chimiques , Organoïdes/métabolisme , Organoïdes/cytologie
15.
Anesth Analg ; 139(2): 385-396, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39008867

RÉSUMÉ

BACKGROUND: Currently, clinical indicators for evaluating endothelial permeability in sepsis are unavailable. Endothelium-derived extracellular vesicles (EDEVs) are emerging as biomarkers of endothelial injury. Platelet endothelial cell adhesion molecule (PECAM) and vascular endothelial (VE)-cadherin are constitutively expressed endothelial intercellular adhesion molecules that regulate intercellular adhesion and permeability. Herein, we investigated the possible association between EDEVs expressing intercellular adhesion molecules (PECAM+ or VE-cadherin+ EDEVs) and endothelial permeability and sepsis severity. METHODS: Human umbilical vein endothelial cells (HUVECs) were stimulated with tumor necrosis factor alpha (TNF-α) directly or after pretreatment with permeability-modifying reagents such as angiopoietin-1, prostacyclin, or vascular endothelial growth factor (VEGF) to alter TNF-α-induced endothelial hyperpermeability. Endothelial permeability was measured using the dextran assay or transendothelial electrical resistance. Additionally, a prospective cross-sectional observational study was conducted to analyze circulating EDEV levels in patients with sepsis. EDEVs were examined in HUVEC culture supernatants or patient plasma (nonsepsis, n = 30; sepsis, n = 30; septic shock, n = 42) using flow cytometry. The Wilcoxon rank-sum test was used for comparisons between 2 groups. Comparisons among 3 or more groups were performed using the Steel-Dwass test. Spearman's test was used for correlation analysis. Statistical significance was set at P < .05. RESULTS: TNF-α stimulation of HUVECs significantly increased EDEV release and endothelial permeability. Pretreatment with angiopoietin-1 or prostacyclin suppressed the TNF-α-induced increase in endothelial permeability and inhibited the release of PECAM+ and VE-cadherin+ EDEVs. In contrast, pretreatment with VEGF increased TNF-α-induced endothelial permeability and the release of PECAM+ and VE-cadherin+ EDEVs. However, pretreatment with permeability-modifying reagents did not affect the release of EDEVs expressing inflammatory stimulus-inducible endothelial adhesion molecules such as E-selectin, intracellular adhesion molecule-1, or vascular cell adhesion molecule-1. The number of PECAM+ EDEVs on admission in the septic-shock group (232 [124, 590]/µL) was significantly higher (P = .043) than that in the sepsis group (138 [77,267]/µL), with an average treatment effect of 98/µL (95% confidence interval [CI], 2-270/µL), and the number of VE-cadherin+ EDEVs in the septic-shock group (173 [76,339]/µL) was also significantly higher (P = .004) than that in the sepsis group (81 [42,159]/µL), with an average treatment effect (ATE) of 79/µL (95% CI, 19-171/µL); these EDEV levels remained elevated until day 5. CONCLUSIONS: EDEVs expressing intercellular adhesion molecules (PECAM+ or VE-cadherin+ EDEVs) may reflect increased endothelial permeability and could be valuable diagnostic and prognostic markers for sepsis.


Sujet(s)
Antigènes CD , Cadhérines , Perméabilité capillaire , Vésicules extracellulaires , Cellules endothéliales de la veine ombilicale humaine , Sepsie , Indice de gravité de la maladie , Humains , Vésicules extracellulaires/métabolisme , Sepsie/métabolisme , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Mâle , Études prospectives , Antigènes CD/métabolisme , Femelle , Adulte d'âge moyen , Cadhérines/métabolisme , Sujet âgé , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Études transversales , Cellules cultivées , Angiopoïétine-1/métabolisme , Marqueurs biologiques/métabolisme , Marqueurs biologiques/sang , Antigènes CD31/métabolisme , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Endothélium vasculaire/métabolisme , Prostacycline/métabolisme
16.
Biochem J ; 481(14): 959-980, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38941070

RÉSUMÉ

While IκB-kinase-ε (IKKε) induces immunomodulatory genes following viral stimuli, its up-regulation by inflammatory cytokines remains under-explored. Since airway epithelial cells respond to airborne insults and potentiate inflammation, IKKε expression was characterized in pulmonary epithelial cell lines (A549, BEAS-2B) and primary human bronchial epithelial cells grown as submersion or differentiated air-liquid interface cultures. IKKε expression was up-regulated by the pro-inflammatory cytokines, interleukin-1ß (IL-1ß) and tumour necrosis factor-α (TNFα). Thus, mechanistic interrogations in A549 cells were used to demonstrate the NF-κB dependence of cytokine-induced IKKε. Furthermore, chromatin immunoprecipitation in A549 and BEAS-2B cells revealed robust recruitment of the NF-κB subunit, p65, to one 5' and two intronic regions within the IKKε locus (IKBKE). In addition, IL-1ß and TNFα induced strong RNA polymerase 2 recruitment to the 5' region, the first intron, and the transcription start site. Stable transfection of the p65-binding regions into A549 cells revealed IL-1ß- and TNFα-inducible reporter activity that required NF-κB, but was not repressed by glucocorticoid. While critical NF-κB motifs were identified in the 5' and downstream intronic regions, the first intronic region did not contain functional NF-κB motifs. Thus, IL-1ß- and TNFα-induced IKKε expression involves three NF-κB-binding regions, containing multiple functional NF-κB motifs, and potentially other mechanisms of p65 binding through non-classical NF-κB binding motifs. By enhancing IKKε expression, IL-1ß may prime, or potentiate, responses to alternative stimuli, as modelled by IKKε phosphorylation induced by phorbol 12-myristate 13-acetate. However, since IKKε expression was only partially repressed by glucocorticoid, IKKε-dependent responses could contribute to glucocorticoid-resistant disease.


Sujet(s)
Cellules épithéliales , I-kappa B Kinase , Humains , I-kappa B Kinase/métabolisme , I-kappa B Kinase/génétique , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules A549 , Facteur de transcription RelA/métabolisme , Facteur de transcription RelA/génétique , Interleukine-1 bêta/pharmacologie , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/génétique , Facteur de transcription NF-kappa B/métabolisme , Facteur de transcription NF-kappa B/génétique , Facteur de nécrose tumorale alpha/pharmacologie , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/génétique , Poumon/métabolisme , Poumon/cytologie , Muqueuse respiratoire/métabolisme , Muqueuse respiratoire/cytologie , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques
17.
Int J Mol Sci ; 25(12)2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38928498

RÉSUMÉ

Extensive evidence supports the connection between obesity-induced inflammation and the heightened expression of IL-6 adipose tissues. However, the mechanism underlying the IL-6 exacerbation in the adipose tissue remains unclear. There is general agreement that TNF-α and stearate concentrations are mildly elevated in adipose tissue in the state of obesity. We hypothesize that TNF-α and stearate co-treatment induce the increased expression of IL-6 in mouse adipocytes. We therefore aimed to determine IL-6 gene expression and protein production by TNF-α/stearate treated adipocytes and investigated the mechanism involved. To test our hypothesis, 3T3-L1 mouse preadipocytes were treated with TNF-α, stearate, or TNF-α/stearate. IL-6 gene expression was assessed by quantitative real-time qPCR. IL-6 protein production secreted in the cell culture media was determined by ELISA. Acetylation of histone was analyzed by Western blotting. Il6 region-associated histone H3 lysine 9/18 acetylation (H3K9/18Ac) was determined by ChIP-qPCR. 3T3-L1 mouse preadipocytes were co-challenged with TNF-α and stearate for 24 h, which led to significantly increased IL-6 gene expression (81 ± 2.1 Fold) compared to controls stimulated with either TNF-α (38 ± 0.5 Fold; p = 0.002) or stearate (56 ± 2.0 Fold; p = 0.013). As expected, co-treatment of adipocytes with TNF-α and stearate significantly increased protein production (338 ± 11 pg/mL) compared to controls stimulated with either TNF-α (28 ± 0.60 pg/mL; p = 0.001) or stearate (53 ± 0.20 pg/mL, p = 0.0015). Inhibition of histone acetyltransferases (HATs) with anacardic acid or curcumin significantly reduced the IL-6 gene expression and protein production by adipocytes. Conversely, TSA-induced acetylation substituted the stimulatory effect of TNF-α or stearate in their synergistic interaction for driving IL-6 gene expression and protein production. Mechanistically, TNF-α/stearate co-stimulation increased the promoter-associated histone H3 lysine 9/18 acetylation (H3K9/18Ac), rendering a transcriptionally permissive state that favored IL-6 expression at the transcriptional and translational levels. Our data represent a TNF-α/stearate cooperativity model driving IL-6 expression in 3T3-L1 cells via the H3K9/18Ac-dependent mechanism, with implications for adipose IL-6 exacerbations in obesity.


Sujet(s)
Cellules 3T3-L1 , Adipocytes , Histone , Interleukine-6 , Facteur de nécrose tumorale alpha , Animaux , Souris , Acétylation , Adipocytes/métabolisme , Adipocytes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Histone/métabolisme , Interleukine-6/métabolisme , Interleukine-6/génétique , Acides stéariques/pharmacologie , Acides stéariques/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie
18.
Immunopharmacol Immunotoxicol ; 46(4): 521-528, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38918176

RÉSUMÉ

OBJECTIVE: Periodontis is a chronic inflammatory disease induced by periodontopathogenic bacteria. The excessive immune response caused by persistent bacterial infection leads to alveolar bone resorption and ultimately tooth loss. Cardamonin is a biologically active substance that is found in the Zingiberaceae family, such as Alpinia zerumbet, and is classified as a natural chalcone. There have been no attempts to use cardamonin for the treatment of periodontitis, and no reports have examined the effects of cardamonin on periodontal tissue component cells. The aim of this study was to analyze effects of cardamonin on expression of inflammation mediators produced by TNFα-stimulated human periodontal ligament cells (HPDLCs), including its effects on signal transduction molecules. METHODS: Cytokine and chemokine levels were measured by ELISA. Protein expression in HPDLCs and activations of signal transduction pathway were determined by Western blotting. RESULTS: Our results indicate that cardamonin suppresses C-C motif chemokine ligand (CCL)2, CCL20, C-X-C motif chemokine ligand (CXCL)10, and interleukin (IL)-6 production and intercellular adhesion molecule (ICAM)-1 and cyclooxygenase (COX)-2 expression in TNF-α-stimulated HPDLCs. In addition, cardamonin induced the expression of the antioxidant enzyme, Heme Oxygenase (HO)-1, in HPDLCs. Furthermore, cardamonin suppressed TNF-α-stimulated c-Jun N-terminal kinase (JNK), nuclear factor (NF)-κB, and signal transducer and activator of transcription (STAT)3 signaling pathways in HPDLCs. CONCLUSION: We show that cardamonin reduces inflammatory mediator production by inhibiting the activation of several signaling pathways in this manuscript.


Sujet(s)
Chalcones , Médiateurs de l'inflammation , Desmodonte , Facteur de nécrose tumorale alpha , Humains , Chalcones/pharmacologie , Desmodonte/effets des médicaments et des substances chimiques , Desmodonte/cytologie , Desmodonte/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Facteur de nécrose tumorale alpha/métabolisme , Médiateurs de l'inflammation/métabolisme , Médiateurs de l'inflammation/antagonistes et inhibiteurs , Cellules cultivées , Transduction du signal/effets des médicaments et des substances chimiques
19.
Biomed Khim ; 70(3): 156-160, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38940204

RÉSUMÉ

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.


Sujet(s)
Vaisseaux coronaires , Doxorubicine , Cellules endothéliales , Interleukine-6 , Humains , Doxorubicine/pharmacologie , Vaisseaux coronaires/cytologie , Vaisseaux coronaires/effets des médicaments et des substances chimiques , Vaisseaux coronaires/métabolisme , Cellules endothéliales/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Interleukine-6/métabolisme , Interleukine-6/génétique , Cellules cultivées , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/pharmacologie , Cytokines/métabolisme , Cytokines/génétique , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Interleukine-8/métabolisme , Interleukine-8/génétique , Chimiokine CCL2/métabolisme , Chimiokine CCL2/génétique , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Interleukine-10/métabolisme , Interleukine-10/génétique
20.
In Vitro Cell Dev Biol Anim ; 60(8): 888-902, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38858305

RÉSUMÉ

Accumulative evidences have indicated the interaction between cellular senescence and ferroptosis. This study intends to investigate the ferroptosis-related molecular markers in TNF-α-induced endothelial senescence. The microarray expression dataset (GSE195517) was used to identify the differently expressed ferroptosis-related genes (DEFRGs) through weighted gene co-expressed network analysis (WGCNA). GO and KEGG were performed to explore the biological function. Furthermore, hub genes were identified after protein-protein interaction (PPI) analysis and validated through real-time qPCR (RT-qPCR). Then, a drug-gene network was established to predict potential drugs for the hub genes. Seven DEFRGs were recognized in the TNF-α-induced HUVEC senescence. Moreover, four hub genes (PTGS2, TNFAIP3, CXCL2, and IL6 are upregulated) were identified by PPI analysis and validated by RT-qPCR. Further analysis exhibited that PTGS2 was subcellularly located in the plasma membrane. Furthermore, after aminosalicylic acid (ASA) was identified as ferroptosis inhibitor for targeting PTGS2 in senescent HUVECs, 5-ASA and 4-ASA were verified to alleviate TNF-α-induced HUVEC senescence through ferroptosis. PTGS2 might play a role in TNF-α-induced HUVEC senescence and ASA may be the potential drug for alleviating TNF-α-induced HUVEC senescence through ferroptosis.


Sujet(s)
Vieillissement de la cellule , Biologie informatique , Cyclooxygenase 2 , Cellules endothéliales de la veine ombilicale humaine , Facteur de nécrose tumorale alpha , Humains , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Facteur de nécrose tumorale alpha/pharmacologie , Facteur de nécrose tumorale alpha/métabolisme , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cyclooxygenase 2/métabolisme , Cyclooxygenase 2/génétique , Ferroptose/effets des médicaments et des substances chimiques , Ferroptose/génétique , Cartes d'interactions protéiques/effets des médicaments et des substances chimiques , Réseaux de régulation génique/effets des médicaments et des substances chimiques , Reproductibilité des résultats
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE