Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.593
Filtrer
1.
PeerJ ; 12: e17862, 2024.
Article de Anglais | MEDLINE | ID: mdl-39135956

RÉSUMÉ

Background: Chemotactic cytokines play a crucial role in the development of acute myeloid leukemia (AML). Thus, investigating the mechanisms of chemotactic cytokine-related genes (CCRGs) in AML is of paramount importance. Methods: Using the TCGA-AML, GSE114868, and GSE12417 datasets, differential expression analysis identified differentially expressed CCRGs (DE-CCRGs). These genes were screened by overlapping differentially expressed genes (DEGs) between AML and control groups with CCRGs. Subsequently, functional enrichment analysis and the construction of a protein-protein interaction (PPI) network were conducted to explore the functions of the DE-CCRGs. Univariate Cox regression, least absolute shrinkage and selection operator (LASSO), and multivariate Cox regression analyses identified relevant prognostic genes and developed a prognostic model. Survival analysis of the prognostic gene was performed, followed by functional similarity analysis, immune analysis, enrichment analysis, and drug prediction analysis. Results: Differential expression analysis revealed 6,743 DEGs, of which 29 DE-CCRGs were selected for this study. Functional enrichment analysis indicated that DE-CCRGs were primarily involved in chemotactic cytokine-related functions and pathways. Six prognostic genes (CXCR3, CXCR2, CXCR6, CCL20, CCL4, and CCR2) were identified and incorporated into the risk model. The model's performance was validated using the GSE12417 dataset. Survival analysis showed significant differences in AML overall survival (OS) between prognostic gene high and low expression groups, indicating that prognostic gene might be significantly associated with patient survival. Additionally, nine different immune cells were identified between the two risk groups. Correlation analysis revealed that CCR2 had the most significant positive correlation with monocytes and the most significant negative correlation with resting mast cells. The tumor immune dysfunction and exclusion score was lower in the high-risk group. Conclusion: CXCR3, CXCR2, CXCR6, CCL20, CCL4, and CCR2 were identified as prognostic genes correlated to AML and the tumor immune microenvironment. These findings offerred novel insights into the prevention and treatment of AML.


Sujet(s)
Leucémie aigüe myéloïde , Cartes d'interactions protéiques , Récepteurs CCR2 , Récepteurs à l'interleukine-8B , Humains , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/mortalité , Pronostic , Récepteurs à l'interleukine-8B/génétique , Récepteurs CCR2/génétique , Cartes d'interactions protéiques/génétique , Chimiokine CCL4/génétique , Chimiokine CCL20/génétique , Chimiokine CCL20/métabolisme , Femelle , Mâle , Chimiokines/génétique , Analyse de profil d'expression de gènes , Adulte d'âge moyen , Marqueurs biologiques tumoraux/génétique , Récepteurs CXCR3
2.
FASEB J ; 38(13): e23757, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38965999

RÉSUMÉ

Hepatic stellate cells (HSCs) are responsible for liver fibrosis accompanied by its activation into myofibroblasts and the abundant production of extracellular matrix. However, the HSC contribution to progression of liver inflammation has been less known. We aimed to elucidate the mechanism in HSCs underlying the inflammatory response and the function of tumor necrosis factor α-related protein A20 (TNFAIP3). We established A20 conditional knockout (KO) mice crossing Twist2-Cre and A20 floxed mice. Using these mice, the effect of A20 was analyzed in mouse liver and HSCs. The human HSC line LX-2 was also used to examine the role and underlying molecular mechanism of A20. In this KO model, A20 was deficient in >80% of HSCs. Spontaneous inflammation with mild fibrosis was found in the liver of the mouse model without any exogenous agents, suggesting that A20 in HSCs suppresses chronic hepatitis. Comprehensive RNA sequence analysis revealed that A20-deficient HSCs exhibited an inflammatory phenotype and abnormally expressed chemokines. A20 suppressed JNK pathway activation in HSCs. Loss of A20 function in LX-2 cells also induced excessive chemokine expression, mimicking A20-deficient HSCs. A20 overexpression suppressed chemokine expression in LX-2. In addition, we identified DCLK1 in the genes regulated by A20. DCLK1 activated the JNK pathway and upregulates chemokine expression. DCLK1 inhibition significantly decreased chemokine induction by A20-silencing, suggesting that A20 controlled chemokine expression in HSCs via the DCLK1-JNK pathway. In conclusion, A20 suppresses chemokine induction dependent on the DCLK1-JNK signaling pathway. These findings demonstrate the therapeutic potential of A20 and the DCLK1-JNK pathway for the regulation of inflammation in chronic hepatitis.


Sujet(s)
Chimiokines , Cellules étoilées du foie , Système de signalisation des MAP kinases , Souris knockout , Protein-Serine-Threonine Kinases , Protéine-3 induite par le facteur de nécrose tumorale alpha , Animaux , Cellules étoilées du foie/métabolisme , Protéine-3 induite par le facteur de nécrose tumorale alpha/métabolisme , Protéine-3 induite par le facteur de nécrose tumorale alpha/génétique , Souris , Humains , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Chimiokines/métabolisme , Chimiokines/génétique , Hépatite chronique/métabolisme , Hépatite chronique/anatomopathologie , Hépatite chronique/génétique , Kinases de type doublecortine , Souris de lignée C57BL , Lignée cellulaire , Mâle
3.
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
4.
Signal Transduct Target Ther ; 9(1): 176, 2024 Jul 22.
Article de Anglais | MEDLINE | ID: mdl-39034318

RÉSUMÉ

Cytokines are critical in regulating immune responses and cellular behavior, playing dual roles in both normal physiology and the pathology of diseases such as cancer. These molecules, including interleukins, interferons, tumor necrosis factors, chemokines, and growth factors like TGF-ß, VEGF, and EGF, can promote or inhibit tumor growth, influence the tumor microenvironment, and impact the efficacy of cancer treatments. Recent advances in targeting these pathways have shown promising therapeutic potential, offering new strategies to modulate the immune system, inhibit tumor progression, and overcome resistance to conventional therapies. In this review, we summarized the current understanding and therapeutic implications of targeting cytokine and chemokine signaling pathways in cancer. By exploring the roles of these molecules in tumor biology and the immune response, we highlighted the development of novel therapeutic agents aimed at modulating these pathways to combat cancer. The review elaborated on the dual nature of cytokines as both promoters and suppressors of tumorigenesis, depending on the context, and discussed the challenges and opportunities this presents for therapeutic intervention. We also examined the latest advancements in targeted therapies, including monoclonal antibodies, bispecific antibodies, receptor inhibitors, fusion proteins, engineered cytokine variants, and their impact on tumor growth, metastasis, and the tumor microenvironment. Additionally, we evaluated the potential of combining these targeted therapies with other treatment modalities to overcome resistance and improve patient outcomes. Besides, we also focused on the ongoing research and clinical trials that are pivotal in advancing our understanding and application of cytokine- and chemokine-targeted therapies for cancer patients.


Sujet(s)
Chimiokines , Cytokines , Tumeurs , Transduction du signal , Humains , Tumeurs/génétique , Tumeurs/immunologie , Tumeurs/traitement médicamenteux , Tumeurs/thérapie , Tumeurs/anatomopathologie , Transduction du signal/immunologie , Transduction du signal/effets des médicaments et des substances chimiques , Cytokines/immunologie , Cytokines/génétique , Cytokines/métabolisme , Chimiokines/immunologie , Chimiokines/génétique , Chimiokines/métabolisme , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Thérapie moléculaire ciblée
5.
PLoS One ; 19(7): e0305816, 2024.
Article de Anglais | MEDLINE | ID: mdl-39038020

RÉSUMÉ

Erythroid cells, serving as progenitors and precursors to erythrocytes responsible for oxygen transport, were shown to exhibit an immunosuppressive and immunoregulatory phenotype. Previous investigations from our research group have revealed an antimicrobial gene expression profile within murine bone marrow erythroid cells which suggested a role for erythroid cells in innate immunity. In the present study, we focused on elucidating the characteristics of human bone marrow erythroid cells through comprehensive analyses, including NanoString gene expression profiling utilizing the Immune Response V2 panel, a BioPlex examination of chemokine and TGF-beta family proteins secretion, and analysis of publicly available single-cell RNA-seq data. Our findings demonstrate that an erythroid cell subpopulation manifests a myeloid-like gene expression signature comprised of antibacterial immunity and neutrophil chemotaxis genes which suggests an involvement of human erythroid cells in the innate immunity. Furthermore, we found that human erythroid cells secreted CCL22, CCL24, CXCL5, CXCL8, and MIF chemokines. The ability of human erythroid cells to express these chemokines might facilitate the restriction of immune cells in the bone marrow under normal conditions or contribute to the ability of erythroid cells to induce local immunosuppression by recruiting immune cells in their immediate vicinity in case of extramedullary hematopoiesis.


Sujet(s)
Cellules érythroïdes , Monocytes , Humains , Monocytes/métabolisme , Monocytes/cytologie , Monocytes/immunologie , Cellules érythroïdes/métabolisme , Cellules érythroïdes/cytologie , Immunité innée , Facteurs inhibiteurs de la migration des macrophages/génétique , Facteurs inhibiteurs de la migration des macrophages/métabolisme , Cellules de la moelle osseuse/métabolisme , Cellules de la moelle osseuse/cytologie , Transcriptome , Analyse de profil d'expression de gènes , Chimiokine CXCL5/métabolisme , Chimiokine CXCL5/génétique , Cellules myéloïdes/métabolisme , Chimiokines/métabolisme , Chimiokines/génétique , Interleukine-8 , Intramolecular oxidoreductases
6.
Int J Mol Sci ; 25(13)2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-39000027

RÉSUMÉ

Cytokines, chemokines, and interferons are released in response to viral infection with the ultimate aim of viral clearance. However, in SARS-CoV-2 infection, there is an imbalanced immune response, with raised cytokine levels but only a limited interferon response with inefficient viral clearance. Furthermore, the inflammatory response can be exaggerated, which risks both acute and chronic sequelae. Several observational studies have suggested a reduced risk of progression to severe COVID-19 in subjects with a higher omega-3 index. However, randomized studies of omega-3 supplementation have failed to replicate this benefit. Omega-3 fats provide important anti-inflammatory effects; however, fatty fish contains many other fatty acids that provide health benefits distinct from omega-3. Therefore, the immune health benefit of whole salmon oil (SO) was assessed in adults with mild to moderate COVID-19. Eleven subjects were randomized to best supportive care (BSC) with or without a full spectrum, enzymatically liberated SO, dosed at 4g daily, for twenty-eight days. Nasal swabs were taken to measure the change in gene expression of markers of immune response and showed that the SO provided both broad inflammation-resolving effects and improved interferon response. The results also suggest improved lung barrier function and enhanced immune memory, although the clinical relevance needs to be assessed in longer-duration studies. In conclusion, the salmon oil was well tolerated and provided broad inflammation-resolving effects, indicating a potential to enhance immune health.


Sujet(s)
COVID-19 , Chimiokines , Cytokines , Huiles de poisson , Interférons , SARS-CoV-2 , Humains , Huiles de poisson/pharmacologie , Huiles de poisson/usage thérapeutique , COVID-19/immunologie , COVID-19/virologie , Mâle , Interférons/métabolisme , Interférons/génétique , SARS-CoV-2/immunologie , Cytokines/métabolisme , Femelle , Adulte d'âge moyen , Chimiokines/métabolisme , Chimiokines/génétique , Adulte , Traitements médicamenteux de la COVID-19 , Acides gras omega-3/pharmacologie
7.
Front Immunol ; 15: 1414891, 2024.
Article de Anglais | MEDLINE | ID: mdl-39076967

RÉSUMÉ

Background: Insect bite hypersensitivity (IBH) is the most frequent skin allergy of horses and is highly debilitating, especially in the chronic phase. IBH is caused by IgE-mediated hypersensitivity reactions to culicoides midge bites and an imbalanced immune response that reduces the welfare of affected horses. Objective: In the present study, we investigated the pathological mechanisms of IBH, aiming to understand the immune cell modulation in acute allergic skin lesions of IBH horses with the goal of finding possible biomarkers for a diagnostic approach to monitor treatment success. Methods: By qPCR, we quantified the gene expression of cytokines, chemokines, and immune receptors in skin punch biopsies of IBH with different severity levels and healthy horses simultaneously in tandem with the analysis of immune cell counts in the blood. Results: Our data show an increase in blood eosinophils, monocytes, and basophils with a concomitant, significant increase in associated cytokine, chemokine, and immune cell receptor mRNA expression levels in the lesional skin of IBH horses. Moreover, IL-5Ra, CCR5, IFN-γ, and IL-31Ra were strongly associated with IBH severity, while IL-31 and IL-33 were rather associated with a milder form of IBH. In addition, our data show a strong correlation of basophil cell count in blood with IL-31Ra, IL-5, IL-5Ra, IFN-γ, HRH2, HRH4, CCR3, CCR5, IL-12b, IL-10, IL-1ß, and CCL26 mRNA expression in skin punch biopsies of IBH horses. Conclusion: In summary, several cytokines and chemokines have been found to be associated with disease severity, hence contributing to IBH pathology. These molecules can be used as potential biomarkers to monitor the onset and progression of the disease or even to evaluate and monitor the efficacy of new therapeutic treatments for IBH skin allergy. To our knowledge, this is the first study that investigated immune cells together with a large set of genes related to their biological function, including correlation to disease severity, in a large cohort of healthy and IBH horses.


Sujet(s)
Chimiokines , Cytokines , Maladies des chevaux , Morsures et piqûres d'insectes , Peau , Animaux , Equus caballus , Morsures et piqûres d'insectes/immunologie , Morsures et piqûres d'insectes/médecine vétérinaire , Peau/immunologie , Peau/anatomopathologie , Maladies des chevaux/immunologie , Chimiokines/génétique , Hypersensibilité/immunologie , Hypersensibilité/médecine vétérinaire , Indice de gravité de la maladie , Ceratopogonidae/immunologie , Mâle , Femelle , Marqueurs biologiques
8.
Genome Res ; 34(6): 837-850, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38977309

RÉSUMÉ

Studies on human parathyroids are generally limited to hyperfunctioning glands owing to the difficulty in obtaining normal human tissue. We therefore obtained non-human primate (NHP) parathyroids to provide a suitable alternative for sequencing that would bear a close semblance to human organs. Single-cell RNA expression analysis of parathyroids from four healthy adult M. mulatta reveals a continuous trajectory of epithelial cell states. Pseudotime analysis based on transcriptomic signatures suggests a progression from GCM2 hi progenitors to mature parathyroid hormone (PTH)-expressing epithelial cells with increasing core mitochondrial transcript abundance along pseudotime. We sequenced, as a comparator, four histologically characterized hyperfunctioning human parathyroids with varying oxyphil and chief cell abundance and leveraged advanced computational techniques to highlight similarities and differences from non-human primate parathyroid expression dynamics. Predicted cell-cell communication analysis reveals abundant endothelial cell interactions in the parathyroid cell microenvironment in both human and NHP parathyroid glands. We show abundant RARRES2 transcripts in both human adenoma and normal primate parathyroid cells and use coimmunostaining to reveal high levels of RARRES2 protein (also known as chemerin) in PTH-expressing cells, which could indicate that RARRES2 plays an unrecognized role in parathyroid endocrine function. The data obtained are the first single-cell RNA transcriptome to characterize nondiseased parathyroid cell signatures and to show a transcriptomic progression of cell states within normal parathyroid glands, which can be used to better understand parathyroid cell biology.


Sujet(s)
Macaca mulatta , Glandes parathyroïdes , Analyse sur cellule unique , Analyse sur cellule unique/méthodes , Humains , Glandes parathyroïdes/métabolisme , Animaux , Transcriptome , Chimiokines/métabolisme , Chimiokines/génétique , Hormone parathyroïdienne/métabolisme , Hormone parathyroïdienne/génétique , Communication cellulaire , Cellules épithéliales/métabolisme , Analyse de profil d'expression de gènes/méthodes , Transcription génétique
9.
FASEB J ; 38(13): e23745, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38923065

RÉSUMÉ

Idiopathic granulomatous mastitis (IGM), a recurrent inflammation disease of the non-lactating breast, has had an increasing clinical morbidity rate in recent years, and its complicated symptoms and unclear etiology make it challenging to treat. This rare benign inflammatory breast disease, centered on the lobules, represents the most challenging type of non-puerperal mastitis (NPM), also known as non-lactating mastitis. In this study, patients diagnosed with IGM (M, n = 23) were recruited as cases, and patients with benign control breast disease (C, n = 17) were enrolled as controls. Cytokine microarray detection measured and analyzed the differentially expressed cytokine factors between IGM and control patients. Then, we verified the mRNA and protein expression levels of the significantly changed cytokine factors using Q-RT-PCR, ELISA, western blot, and IHC experiments. The cytokine factor expression levels significantly changed compared to the control group. We observed a significant increase between IGM and control patients in cytokine factors expression, such as interleukin-1ß (IL-1ß), monokine induced by gamma interferon (MIG), macrophage inflammatory protein (MIP)-1α, MIP-1ß, tumor necrosis factor receptor 2 (TNF RII). Then, we verified the expression of these top five dysregulated factors in both mRNA and protein levels. Our results demonstrated the cytokine map in IGM and indicated that several cytokines, especially chemokines, were associated with and significantly dysregulated in IGM tissues compared to the control group. The chemokine factors involved might be essential in developing and treating IGM. These findings would be helpful for a better understanding of IGM and offer valuable insights for devising novel diagnostic and therapeutic strategies.


Sujet(s)
Chimiokines , Mastite granulomateuse , Humains , Femelle , Mastite granulomateuse/métabolisme , Mastite granulomateuse/génétique , Adulte , Chimiokines/métabolisme , Chimiokines/génétique , Adulte d'âge moyen , Cytokines/métabolisme , Cytokines/génétique , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/génétique , Études cas-témoins , Chimiokine CXCL9/métabolisme , Chimiokine CXCL9/génétique
10.
Toxins (Basel) ; 16(6)2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38922170

RÉSUMÉ

Snakebite envenoming is a neglected tropical disease that causes >100,000 deaths and >400,000 cases of morbidity annually. Despite the use of mouse models, severe local envenoming, defined by morbidity-causing local tissue necrosis, remains poorly understood, and human-tissue responses are ill-defined. Here, for the first time, an ex vivo, non-perfused human skin model was used to investigate temporal histopathological and immunological changes following subcutaneous injections of venoms from medically important African vipers (Echis ocellatus and Bitis arietans) and cobras (Naja nigricollis and N. haje). Histological analysis of venom-injected ex vivo human skin biopsies revealed morphological changes in the epidermis (ballooning degeneration, erosion, and ulceration) comparable to clinical signs of local envenoming. Immunostaining of these biopsies confirmed cell apoptosis consistent with the onset of necrosis. RNA sequencing, multiplex bead arrays, and ELISAs demonstrated that venom-injected human skin biopsies exhibited higher rates of transcription and expression of chemokines (CXCL5, MIP1-ALPHA, RANTES, MCP-1, and MIG), cytokines (IL-1ß, IL-1RA, G-CSF/CSF-3, and GM-CSF), and growth factors (VEGF-A, FGF, and HGF) in comparison to non-injected biopsies. To investigate the efficacy of antivenom, SAIMR Echis monovalent or SAIMR polyvalent antivenom was injected one hour following E. ocellatus or N. nigricollis venom treatment, respectively, and although antivenom did not prevent venom-induced dermal tissue damage, it did reduce all pro-inflammatory chemokines, cytokines, and growth factors to normal levels after 48 h. This ex vivo skin model could be useful for studies evaluating the progression of local envenoming and the efficacy of snakebite treatments.


Sujet(s)
Cytokines , Nécrose , Peau , Humains , Peau/anatomopathologie , Peau/effets des médicaments et des substances chimiques , Animaux , Cytokines/métabolisme , Cytokines/génétique , Morsures de serpent/anatomopathologie , Venins des élapidés/toxicité , Venins de vipère/toxicité , Inflammation/anatomopathologie , Inflammation/induit chimiquement , Viperidae , Chimiokines/métabolisme , Chimiokines/génétique
11.
Cell Rep Med ; 5(7): 101622, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38917802

RÉSUMÉ

Progressive multifocal leukoencephalopathy (PML) has been associated with different forms of immune compromise. This study analyzes the chemokine signals and attracted immune cells in cerebrospinal fluid (CSF) during PML to define immune cell subpopulations relevant for the PML immune response. In addition to chemokines that indicate a general state of inflammation, like CCL5 and CXCL10, the CSF of PML patients specifically contains CCL2 and CCL4. Single-cell transcriptomics of CSF cells suggests an enrichment of distinct CD4+ and CD8+ T cells expressing chemokine receptors CCR2, CCR5, and CXCR3, in addition to ITGA4 and the genetic PML risk genes STXBP2 and LY9. This suggests that specific immune cell subpopulations migrate into the central nervous system to mitigate PML, and their absence might coincide with PML development. Monitoring them might hold clues for PML risk, and boosting their recruitment or function before therapeutic immune reconstitution might improve its risk-benefit ratio.


Sujet(s)
Mouvement cellulaire , Système nerveux central , Chimiokines , Leucoencéphalopathie multifocale progressive , Humains , Leucoencéphalopathie multifocale progressive/anatomopathologie , Leucoencéphalopathie multifocale progressive/immunologie , Chimiokines/métabolisme , Chimiokines/génétique , Mouvement cellulaire/génétique , Système nerveux central/anatomopathologie , Système nerveux central/métabolisme , Système nerveux central/immunologie , Lymphocytes T CD8+/immunologie , Mâle , Femelle , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Adulte d'âge moyen , Sujet âgé
12.
Int J Mol Sci ; 25(10)2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38791448

RÉSUMÉ

Chemokines are key proteins that regulate cell migration and immune responses and are essential for modulating the tumor microenvironment. Despite their close association with colon cancer, the expression patterns, prognosis, immunity, and specific roles of chemokines in colon cancer are still not fully understood. In this study, we investigated the mutational features, differential expression, and immunological characteristics of chemokines in colon cancer (COAD) by analyzing the Tumor Genome Atlas (TCGA) database. We clarified the biological functions of these chemokines using Gene Ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. By univariate and multivariate COX regression analyses, we developed chemokine-based prognostic risk models. In addition, using Gene Set Enrichment Analysis (GSEA) and Gene Set Variant Analysis (GSVA), we analyzed the differences in immune responses and signaling pathways among different risk groups. The results showed that the mutation rate of chemokines was low in COAD, but 25 chemokines were significantly differentially expressed. These chemokines function in several immune-related biological processes and play key roles in signaling pathways including cytokine-cytokine receptor interactions, NF-kappa B, and IL-17. Prognostic risk models based on CCL22, CXCL1, CXCL8, CXCL9, and CXCL11 performed well. GSEA and GSVA analyses showed significant differences in immune responses and signaling pathways across risk groups. In conclusion, this study reveals the potential molecular mechanisms of chemokines in COAD and proposes a new prognostic risk model based on these insights.


Sujet(s)
Chimiokines , Tumeurs du côlon , Humains , Chimiokines/génétique , Chimiokines/métabolisme , Tumeurs du côlon/génétique , Tumeurs du côlon/immunologie , Pronostic , Régulation de l'expression des gènes tumoraux , Mutation , Transduction du signal , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Gene Ontology , Femelle , Mâle , Bases de données génétiques , Marqueurs biologiques tumoraux/génétique , Analyse de profil d'expression de gènes
13.
Cell ; 187(13): 3390-3408.e19, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38754421

RÉSUMÉ

Clinical trials have identified ARID1A mutations as enriched among patients who respond favorably to immune checkpoint blockade (ICB) in several solid tumor types independent of microsatellite instability. We show that ARID1A loss in murine models is sufficient to induce anti-tumor immune phenotypes observed in ARID1A mutant human cancers, including increased CD8+ T cell infiltration and cytolytic activity. ARID1A-deficient cancers upregulated an interferon (IFN) gene expression signature, the ARID1A-IFN signature, associated with increased R-loops and cytosolic single-stranded DNA (ssDNA). Overexpression of the R-loop resolving enzyme, RNASEH2B, or cytosolic DNase, TREX1, in ARID1A-deficient cells prevented cytosolic ssDNA accumulation and ARID1A-IFN gene upregulation. Further, the ARID1A-IFN signature and anti-tumor immunity were driven by STING-dependent type I IFN signaling, which was required for improved responsiveness of ARID1A mutant tumors to ICB treatment. These findings define a molecular mechanism underlying anti-tumor immunity in ARID1A mutant cancers.


Sujet(s)
Lymphocytes T CD8+ , Protéines de liaison à l'ADN , Interféron de type I , Protéines membranaires , Tumeurs , Transduction du signal , Facteurs de transcription , Animaux , Humains , Souris , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Lignée cellulaire tumorale , Protéines de liaison à l'ADN/métabolisme , Exodeoxyribonucleases/métabolisme , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Interféron de type I/métabolisme , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Souris de lignée C57BL , Mutation , Tumeurs/immunologie , Tumeurs/génétique , Protéines nucléaires/métabolisme , Phosphoprotéines/métabolisme , Facteurs de transcription/métabolisme , Mâle , Chimiokines/génétique , Chimiokines/métabolisme
14.
Comput Biol Med ; 177: 108666, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38820773

RÉSUMÉ

BACKGROUND: α-1,3-mannosyltransferase (ALG3) holds significance as a key member within the mannosyltransferase family. Nevertheless, the exact function of ALG3 in cancer remains ambiguous. Consequently, the current research aimed to examine the function and potential mechanisms of ALG3 in various types of cancer. METHODS: Deep pan-cancer analyses were conducted to investigate the expression patterns, prognostic value, genetic variations, single-cell omics, immunology, and drug responses associated with ALG3. Subsequently, in vitro experiments were executed to ascertain the biological role of ALG3 in breast cancer. Moreover, the link between ALG3 and CD8+ T cells was verified using immunofluorescence. Lastly, the association between ALG3 and chemokines was assessed using qRT-PCR and ELISA. RESULTS: Deep pan-cancer analysis demonstrated a heightened expression of ALG3 in the majority of tumors based on multi-omics evidence. ALG3 emerges as a diagnostic and prognostic biomarker across diverse cancer types. In addition, ALG3 participates in regulating the tumor immune microenvironment. Elevated levels of ALG3 were closely linked to the infiltration of bone marrow-derived suppressor cells (MDSC) and CD8+ T cells. According to in vitro experiments, ALG3 promotes proliferation and migration of breast cancer cells. Moreover, ALG3 inhibited CD8+ T cell infiltration by suppressing chemokine secretion. Finally, the inhibition of ALG3 enhanced the responsiveness of breast cancer cells to 5-fluorouracil treatment. CONCLUSION: ALG3 shows potential as both a prognostic indicator and immune infiltration biomarker across various types of cancer. Inhibition of ALG3 may represent a promising therapeutic strategy for tumor treatment.


Sujet(s)
Lymphocytes T CD8+ , Fluorouracil , Humains , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/métabolisme , Fluorouracil/pharmacologie , Chimiokines/métabolisme , Chimiokines/génétique , Femelle , Lignée cellulaire tumorale , Tumeurs/traitement médicamenteux , Tumeurs/immunologie , Tumeurs/génétique , Tumeurs/métabolisme , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/immunologie , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Multi-omique
15.
Sci Rep ; 14(1): 10610, 2024 05 09.
Article de Anglais | MEDLINE | ID: mdl-38719857

RÉSUMÉ

Histone lysine methylation is thought to play a role in the pathogenesis of rheumatoid arthritis (RA). We previously reported aberrant expression of the gene encoding mixed-lineage leukemia 1 (MLL1), which catalyzes methylation of histone H3 lysine 4 (H3K4), in RA synovial fibroblasts (SFs). The aim of this study was to elucidate the involvement of MLL1 in the activated phenotype of RASFs. SFs were isolated from synovial tissues obtained from patients with RA or osteoarthritis (OA) during total knee joint replacement. MLL1 mRNA and protein levels were determined after stimulation with tumor necrosis factor α (TNFα). We also examined changes in trimethylation of H3K4 (H3K4me3) levels in the promoters of RA-associated genes (matrix-degrading enzymes, cytokines, and chemokines) and the mRNA levels upon small interfering RNA-mediated depletion of MLL1 in RASFs. We then determined the levels of H3K4me3 and mRNAs following treatment with the WD repeat domain 5 (WDR5)/MLL1 inhibitor MM-102. H3K4me3 levels in the gene promoters were also compared between RASFs and OASFs. After TNFα stimulation, MLL1 mRNA and protein levels were higher in RASFs than OASFs. Silencing of MLL1 significantly reduced H3K4me3 levels in the promoters of several cytokine (interleukin-6 [IL-6], IL-15) and chemokine (C-C motif chemokine ligand 2 [CCL2], CCL5, C-X-C motif chemokine ligand 9 [CXCL9], CXCL10, CXCL11, and C-X3-C motif chemokine ligand 1 [CX3CL1]) genes in RASFs. Correspondingly, the mRNA levels of these genes were significantly decreased. MM-102 significantly reduced the promoter H3K4me3 and mRNA levels of the CCL5, CXCL9, CXCL10, and CXCL11 genes in RASFs. In addition, H3K4me3 levels in the promoters of the IL-6, IL-15, CCL2, CCL5, CXCL9, CXCL10, CXCL11, and CX3CL1 genes were significantly higher in RASFs than OASFs. Our findings suggest that MLL1 regulates the expression of particular cytokines and chemokines in RASFs and is associated with the pathogenesis of RA. These results could lead to new therapies for RA.


Sujet(s)
Polyarthrite rhumatoïde , Histone-lysine N-methyltransferase , Protéine de la leucémie myéloïde-lymphoïde , Membrane synoviale , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/génétique , Cellules cultivées , Chimiokines/métabolisme , Chimiokines/génétique , Cytokines/métabolisme , Fibroblastes/métabolisme , Régulation de l'expression des gènes , Histone-lysine N-methyltransferase/métabolisme , Histone-lysine N-methyltransferase/génétique , Histone/métabolisme , Protéine de la leucémie myéloïde-lymphoïde/métabolisme , Protéine de la leucémie myéloïde-lymphoïde/génétique , Arthrose/métabolisme , Arthrose/anatomopathologie , Arthrose/génétique , Régions promotrices (génétique) , ARN messager/métabolisme , ARN messager/génétique , Membrane synoviale/métabolisme , Membrane synoviale/anatomopathologie , Facteur de nécrose tumorale alpha/métabolisme
16.
Int Immunopharmacol ; 134: 112172, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38703566

RÉSUMÉ

BACKGROUND: The clinical significance and comprehensive characteristics of chemokines and chemokine receptors in colorectal cancer (CRC) have not been previously reported. Our study aims to investigate the expression profiles of chemokines and chemokine receptors, as well as establish subtypes in CRC. METHODS: 1009 CRC samples were enrolled in our study. Consensus unsupervised clustering analysis was conducted to establish subtypes, and a risk score model was developed using univariate Cox regression and least absolute shrinkage and selection operator (LASSO) analyses. 36 pairs of tissue specimens of CRC patients and two CRC cell lines were used to validate the subtypes and risk score in vitro. Quantitative real-time PCR and western blotting were employed to validate mRNA and protein expression levels, respectively. Flow cytometry was utilized for analyzing cell apoptosis, while cell viability assay and EdU assay were conducted to assess cell proliferation ability. RESULTS: The Cluster B group shares similarities with the low-risk group in terms of exhibiting a higher level of immune cell infiltration and belonging to hot tumor. Patients CRC in the Cluster B group demonstrate a more favorable prognosis and exhibit better response to immunotherapy and chemotherapy. On the other hand, the Cluster A group resembles the high-risk group as it displays lower levels of immune cell infiltration, indicating a cold tumor phenotype. CRC patients in the Cluster A group have poorer prognoses and show less therapeutic efficacy towards immunotherapy and chemotherapy. Furthermore, we utilized a total of 36 pairs of tissue samples obtained from patients with CRC, along with two CRC cell lines for validation in vitro. This comprehensive approach further enhances the scientific validity and reliability of the identified subtypes and risk score in their ability to predict prognosis, response to immunotherapy, and response to chemotherapy among CRC patients. CONCLUSION: We first established robust prognostic subtypes based on chemokines and chemokine receptors, which could potentially serve as a novel biomarker for guiding individualized treatment in patients with CRC undergoing immunotherapy and chemotherapy.


Sujet(s)
Chimiokines , Tumeurs colorectales , Immunothérapie , Récepteurs aux chimiokines , Humains , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/immunologie , Tumeurs colorectales/diagnostic , Tumeurs colorectales/thérapie , Immunothérapie/méthodes , Pronostic , Femelle , Mâle , Chimiokines/métabolisme , Chimiokines/génétique , Récepteurs aux chimiokines/métabolisme , Récepteurs aux chimiokines/génétique , Adulte d'âge moyen , Marqueurs biologiques tumoraux/métabolisme , Lignée cellulaire tumorale , Sujet âgé , Régulation de l'expression des gènes tumoraux , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques
17.
Sci Rep ; 14(1): 7672, 2024 04 01.
Article de Anglais | MEDLINE | ID: mdl-38561377

RÉSUMÉ

Lipopolysaccharide (LPS) is known to elicit a robust immune response. This study aimed to investigate the impact of LPS on the transcriptome of human nasal epithelial cells (HNEpC). HNEpC were cultured and stimulated with LPS (1 µg/mL) or an equivalent amount of normal culture medium. Subsequently, total RNA was extracted, purified, and sequenced using next-generation RNA sequencing technology. Differentially expressed genes (DEGs) were identified and subjected to functional enrichment analysis. A protein-protein interaction (PPI) network of DEGs was constructed, followed by Ingenuity Pathway Analysis (IPA) to identify molecular pathways influenced by LPS exposure on HNEpC. Validation of key genes was performed using quantitative real-time PCR (qRT-PCR). A total of 97 DEGs, comprising 48 up-regulated genes and 49 down-regulated genes, were identified. Results from functional enrichment analysis, PPI, and IPA indicated that DEGs were predominantly enriched in chemokine-related signaling pathways. Subsequent qRT-PCR validation demonstrated significant upregulation of key genes in these pathways in LPS-treated HNEpC compared to control cells. In conclusion, LPS intervention profoundly altered the transcriptome of HNEpC, potentially exacerbating inflammatory responses through the activation of chemokine-related signaling pathways.


Sujet(s)
Analyse de profil d'expression de gènes , Lipopolysaccharides , Humains , Analyse de profil d'expression de gènes/méthodes , Lipopolysaccharides/pharmacologie , Transcriptome , Transduction du signal/génétique , Cellules épithéliales , Chimiokines/génétique , Biologie informatique/méthodes
18.
Sci Rep ; 14(1): 8196, 2024 04 08.
Article de Anglais | MEDLINE | ID: mdl-38589444

RÉSUMÉ

In atherosclerotic lesions, monocyte-derived macrophages are major source of interferon gamma (IFN-γ), a pleotropic cytokine known to regulate the expression of numerous genes, including the antiviral gene RSAD2. While RSAD2 was reported to be expressed in endothelial cells of human carotid lesions, its significance for the development of atherosclerosis remains utterly unknown. Here, we harnessed publicly available human carotid atherosclerotic data to explore RSAD2 in lesions and employed siRNA-mediated gene-knockdown to investigate its function in IFN-γ-stimulated human aortic smooth muscle cells (hAoSMCs). Silencing RSAD2 in IFN-γ-stimulated hAoSMCs resulted in reduced expression and secretion of key CXCR3-chemokines, CXCL9, CXCL10, and CXCL11. Conditioned medium from RSAD2-deficient hAoSMCs exhibited diminished monocyte attraction in vitro compared to conditioned medium from control cells. Furthermore, RSAD2 transcript was elevated in carotid lesions where it was expressed by several different cell types, including endothelial cells, macrophages and smooth muscle cells. Interestingly, RSAD2 displayed significant correlations with CXCL10 (r = 0.45, p = 0.010) and CXCL11 (r = 0.53, p = 0.002) in human carotid lesions. Combining our findings, we uncover a novel role for RSAD2 in hAoSMCs, which could potentially contribute to monocyte recruitment in the context of atherosclerosis.


Sujet(s)
Athérosclérose , Plaque d'athérosclérose , Humains , Plaque d'athérosclérose/génétique , Interférons , Cellules endothéliales/métabolisme , Milieux de culture conditionnés/pharmacologie , Chimiokines/génétique , Chimiokines/métabolisme , Chimiokine CXCL11/génétique , Chimiokine CXCL11/métabolisme , Chimiokine CXCL9/métabolisme , Interféron gamma/pharmacologie , Interféron gamma/métabolisme , Athérosclérose/génétique , Myocytes du muscle lisse/métabolisme , Chimiokine CXCL10/génétique , Chimiokine CXCL10/métabolisme , Récepteurs CXCR3/génétique , Récepteurs CXCR3/métabolisme , Protéine Viperin
19.
PLoS One ; 19(4): e0293680, 2024.
Article de Anglais | MEDLINE | ID: mdl-38652715

RÉSUMÉ

Universal and early recognition of pathogens occurs through recognition of evolutionarily conserved pathogen associated molecular patterns (PAMPs) by innate immune receptors and the consequent secretion of cytokines and chemokines. The intrinsic complexity of innate immune signaling and associated signal transduction challenges our ability to obtain physiologically relevant, reproducible and accurate data from experimental systems. One of the reasons for the discrepancy in observed data is the choice of measurement strategy. Immune signaling is regulated by the interplay between pathogen-derived molecules with host cells resulting in cellular expression changes. However, these cellular processes are often studied by the independent assessment of either the transcriptome or the proteome. Correlation between transcription and protein analysis is lacking in a variety of studies. In order to methodically evaluate the correlation between transcription and protein expression profiles associated with innate immune signaling, we measured cytokine and chemokine levels following exposure of human cells to the PAMP lipopolysaccharide (LPS) from the Gram-negative pathogen Pseudomonas aeruginosa. Expression of 84 messenger RNA (mRNA) transcripts and 69 proteins, including 35 overlapping targets, were measured in human lung epithelial cells. We evaluated 50 biological replicates to determine reproducibility of outcomes. Following pairwise normalization, 16 mRNA transcripts and 6 proteins were significantly upregulated following LPS exposure, while only five (CCL2, CSF3, CXCL5, CXCL8/IL8, and IL6) were upregulated in both transcriptomic and proteomic analysis. This lack of correlation between transcription and protein expression data may contribute to the discrepancy in the immune profiles reported in various studies. The use of multiomic assessments to achieve a systems-level understanding of immune signaling processes can result in the identification of host biomarker profiles for a variety of infectious diseases and facilitate countermeasure design and development.


Sujet(s)
Marqueurs biologiques , Cellules épithéliales , Lipopolysaccharides , Pseudomonas aeruginosa , Humains , Lipopolysaccharides/pharmacologie , Cellules épithéliales/métabolisme , Cellules épithéliales/immunologie , Pseudomonas aeruginosa/immunologie , Marqueurs biologiques/métabolisme , Poumon/métabolisme , Poumon/immunologie , Transcriptome , Cytokines/métabolisme , Analyse de profil d'expression de gènes , Immunité innée , ARN messager/génétique , ARN messager/métabolisme , Transcription génétique/effets des médicaments et des substances chimiques , Chimiokines/métabolisme , Chimiokines/génétique
20.
J Ethnopharmacol ; 332: 118245, 2024 Oct 05.
Article de Anglais | MEDLINE | ID: mdl-38679399

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: The process of atherosclerosis (AS) is complicated. Transcriptomics technology can assist in discovering the underlying mechanisms and exploring the key targets of Traditional Chinese Medicine (TCM) against atherosclerosis. AIM: This study aimed to investigate targets and signaling pathways significantly related to AS and the potential intervention targets of Xuefu Zhuyu decoction by transcriptomics. MATERIALS AND METHODS: AS models were established by subjecting ApoE-/-mice to an 8-week high-fat diet. Structural changes and plaque formation in the aortic root were observed using hematoxylin-eosin staining (HE staining), while Oil Red O staining was employed to visualize lipid deposition within the aortic root plaque. Movat staining and immunohistochemical staining were conducted to examine the components present in the aortic root plaque. Macrophage content within the plaque was observed through immunofluorescence. Additionally, mRNA sequencing was performed on aortic tissues to identify differentially expressed genes. Enrichment analysis was performed using GO and KEGG analysis. Visualization of the protein-protein interaction (PPI) network was achieved using Cytoscape 3.7.1 and STRING. Western blotting (WB) was employed to assess the protein expression of major differentially expressed genes in the aortic tissue. The drug freeze-dried powder of Xuefu Zhuyu decoction was prepared and the RAW264.7 cells were induced by lipopolysaccharide (LPS) to build an in vitro model. Real-time quantitative PCR was employed to measure the mRNA expression of major differential genes. RESULTS: After ApoE-/- mice were fed with an 8-week high-fat diet, observable changes included the thinning of the aortic root wall, the accumulation of foam cells within the plaque, and the formation of cholesterol crystals in the model group. Treatment with Xuefu Zhuyu (XFZY) decoction for 12 weeks significantly reduced the lipid deposition and the number of macrophages (P < 0.05) and significantly increased the collagen content within the plaque (P < 0.01). Enrichment analysis revealed a high enrichment of the Cytokine-cytokine receptor interaction pathway and Chemokine signaling pathway. Noteworthy genes involved in this response included Ccl12, Ccl22, Cx3cr1, Ccr7, Ccr2, Tnfrsf25, and Gdf5. Xuefu Zhuyu decoction significantly downregulated the expression of CX3CL1 and CX3CR1 (P < 0.05) and upregulated the expression of GDF5 (P < 0.01). Compared with control group, in cell models, the mRNA expressions of Ccl12, Ccl22, and Ccr2 were significantly upregulated (P < 0.05 or P < 0.01). Xuefu Zhuyu decoction significantly downregulated the expression of Ccl12, Ccl22, Cx3cr1, Ccr7 and Ccr2 (P < 0.05 or P < 0.01). CONCLUSION: Xuefu Zhuyu decoction demonstrates effective regulation of plaque components, retarding plaque progression and preserving plaque stability by modulating lipid metabolism and inflammatory responses. Subsequent transcriptome analysis identified the Cytokine-cytokine receptor interaction and Chemokine signaling pathway as potential key pathways for the therapeutic effects of Xuefu Zhuyu decoction. This insight not only provides crucial avenues for further exploration into the mechanisms underlying Xuefu Zhuyu decoction but also offers valuable perspectives and hypotheses for enhancing disease prevention and treatment strategies.


Sujet(s)
Athérosclérose , Alimentation riche en graisse , Médicaments issus de plantes chinoises , Transduction du signal , Animaux , Médicaments issus de plantes chinoises/pharmacologie , Athérosclérose/traitement médicamenteux , Athérosclérose/métabolisme , Souris , Transduction du signal/effets des médicaments et des substances chimiques , Mâle , Alimentation riche en graisse/effets indésirables , Chimiokines/métabolisme , Chimiokines/génétique , Analyse de profil d'expression de gènes/méthodes , Souris invalidées pour les gènes ApoE , Souris de lignée C57BL , Plaque d'athérosclérose/traitement médicamenteux , Modèles animaux de maladie humaine , Transcriptome/effets des médicaments et des substances chimiques , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Apolipoprotéines E/génétique , Aorte/effets des médicaments et des substances chimiques , Aorte/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE