Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 20.360
Filtrer
1.
Front Immunol ; 15: 1437046, 2024.
Article de Anglais | MEDLINE | ID: mdl-39156888

RÉSUMÉ

Metabolic dysfunction-associated steatotic liver disease (MASLD) comprises a spectrum of liver diseases that span simple steatosis, metabolic dysfunction-associated steatohepatitis (MASH) and fibrosis and may progress to cirrhosis and cancer. The pathogenesis of MASLD is multifactorial and is driven by environmental, genetic, metabolic and immune factors. This review will focus on the role of the type 3 cytokines IL-17 and IL-22 in MASLD pathogenesis and progression. IL-17 and IL-22 are produced by similar adaptive and innate immune cells such as Th17 and innate lymphoid cells, respectively. IL-17-related signaling is upregulated during MASLD resulting in increased chemokines and proinflammatory cytokines in the liver microenvironment, enhanced recruitment of myeloid cells and T cells leading to exacerbation of inflammation and liver disease progression. IL-17 may also act directly by activating hepatic stellate cells resulting in increased fibrosis. In contrast, IL-22 is a pleiotropic cytokine with a dominantly protective signature in MASLD and is currently being tested as a therapeutic strategy. IL-22 also exhibits beneficial metabolic effects and abrogates MASH-related inflammation and fibrosis development via inducing the production of anti-oxidants and anti-apoptotic factors. A sex-dependent effect has been attributed to both cytokines, most importantly to IL-22 in MASLD or related conditions. Altogether, IL-17 and IL-22 are key effectors in MASLD pathogenesis and progression. We will review the role of these two cytokines and cells that produce them in the development of MASLD, their interaction with host factors driving MASLD including sexual dimorphism, and their potential therapeutic benefits.


Sujet(s)
Interleukine-17 , , Interleukines , Humains , Interleukine-17/métabolisme , Interleukine-17/immunologie , Interleukines/métabolisme , Interleukines/immunologie , Animaux , Stéatose hépatique/immunologie , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Maladies métaboliques/métabolisme , Maladies métaboliques/immunologie , Foie/anatomopathologie , Foie/métabolisme , Foie/immunologie
2.
AIDS ; 38(11): 1725-1727, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39088830

RÉSUMÉ

We explored the impact of immune dysregulation on pancreatic beta cell injury in HIV patients. Analyzing 105 participant samples, we observed lower IL-21 levels and elevated immune checkpoint levels (e.g. PD-1, CD27+, CD40+) in untreated HIV patients. Notably, soluble TIM-3 correlated positively with improved beta cell function and inversely with beta cell stress, suggesting its potential role in beta cell protection in untreated HIV.


Sujet(s)
Infections à VIH , Cellules à insuline , Humains , Infections à VIH/complications , Infections à VIH/immunologie , Cellules à insuline/immunologie , Mâle , Femelle , Adulte d'âge moyen , Adulte , Récepteur cellulaire-2 du virus de l'hépatite A , Interleukines/sang , Protéines de points de contrôle immunitaires/métabolisme
3.
Clin Exp Med ; 24(1): 184, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39117877

RÉSUMÉ

The prevalence of HCV infection in Egypt has decreased following the introduction of direct-acting antiviral therapy. However, treatment response is influenced by various factors, particularly host immunogenetics such as IL-28B and FOXP3 polymorphisms. The current study examined the impact of SNPs in the FOXP3 gene promoter region on HCV-infected Egyptian patients, along with SNPs in the IL28B gene.This study involved 99 HCV patients who achieved SVR12 after a 12 week DAA treatment while 63 HCV patients experienced treatment failure. IL28B rs12979860 SNP was identified using real-time PCR, while IL28B rs8099917, FOXP3 rs3761548, and rs2232365 SNPs were analyzed using RFLP-PCR. Serum levels of IL28B and FOXP3 were quantified using ELISA technique in representative samples from both groups. The IL28B rs12979860 T > C (P = 0.013) and FOXP3 rs2232365 A > G polymorphisms (P = 0.008) were found to significantly increase the risk of non-response. Responders had higher IL28B serum levels (P = 0.046) and lower FOXP3 levels (P < 0.001) compared to non-responders. Regression analysis showed an association between IL28B rs12979860 and FOXP3 rs2232365 with treatment response, independent of age and gender. A predictive model was developed with 76.2% sensitivity and 91.9% specificity for estimating DAAs response in HCV patients.Our findings confirmed the IL28B rs12979860 T > C and FOXP3 rs2232365 A > G polymorphisms significantly affect DAA treatment response in HCV Egyptian patients. Lower levels of IL-28B along with higher levels of FOXP3 are linked to poor response. Our results may lead to new insights into DAA responsiveness contributing to personalized medicine and improving therapeutic decision-making for HCV patients.


Sujet(s)
Antiviraux , Facteurs de transcription Forkhead , Hépatite C chronique , Interférons , Interleukines , Polymorphisme de nucléotide simple , Humains , Hépatite C chronique/traitement médicamenteux , Hépatite C chronique/génétique , Hépatite C chronique/virologie , Mâle , Femelle , Antiviraux/usage thérapeutique , Adulte d'âge moyen , Interleukines/génétique , Interleukines/sang , Adulte , Égypte , Facteurs de transcription Forkhead/génétique , Résultat thérapeutique , Régions promotrices (génétique) , Immunogénétique , Interféron lambda
4.
J Neuroinflammation ; 21(1): 198, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39118084

RÉSUMÉ

Astrocytes respond and contribute to neuroinflammation by adopting inflammatory reactive states. Although recent efforts have characterized the gene expression signatures associated with these reactive states, the cell biology underlying inflammatory reactive astrocyte phenotypes remains under-explored. Here, we used CRISPR-based screening in human iPSC-derived astrocytes to identify mTOR activation a driver of cytokine-induced endolysosomal system remodeling, manifesting as alkalinization of endolysosomal compartments, decreased autophagic flux, and increased exocytosis of certain endolysosomal cargos. Through endolysosomal proteomics, we identified and focused on one such cargo-IL-32, a disease-associated pro-inflammatory cytokine not present in rodents, whose secretion mechanism is not well understood. We found that IL-32 was partially secreted in extracellular vesicles likely to be exosomes. Furthermore, we found that IL-32 was involved in the polarization of inflammatory reactive astrocyte states and was upregulated in astrocytes in multiple sclerosis lesions. We believe that our results advance our understanding of cell biological pathways underlying inflammatory reactive astrocyte phenotypes and identify potential therapeutic targets.


Sujet(s)
Astrocytes , Exosomes , Interleukines , Lysosomes , Sérine-thréonine kinases TOR , Astrocytes/métabolisme , Humains , Exosomes/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Lysosomes/métabolisme , Interleukines/métabolisme , Endosomes/métabolisme , Cellules souches pluripotentes induites/métabolisme , Cellules cultivées , Maladies neuro-inflammatoires/métabolisme , Maladies neuro-inflammatoires/anatomopathologie , Inflammation/métabolisme , Inflammation/anatomopathologie
5.
Front Immunol ; 15: 1359481, 2024.
Article de Anglais | MEDLINE | ID: mdl-39104539

RÉSUMÉ

Generalized pustular psoriasis (GPP) is a chronic, rare, and potentially life-threatening inflammatory disease, characterized by the rapid and widespread eruption of small, sterile pustules with surrounding skin erythema. Abnormal signaling of the interleukin-36 (IL-36) pathway appears to have a central role in GPP immunopathology, and provides a rational therapeutic target. Spesolimab is a first-in-class humanized monoclonal antibody that binds specifically to the IL-36 receptor, and antagonizes IL-36 signaling. Spesolimab obtained regulatory approval in the United States (US) in September 2022 for use in the treatment of GPP flares in adults, and was subsequently approved for GPP flare treatment in many other countries across the world. Recently, regulatory approval was granted for subcutaneous dosing of spesolimab for treatment of GPP when not experiencing a flare. Here, we review data from two key clinical trials that supported the initial US regulatory approval; namely, the phase 1 proof-of-concept trial (ClinicalTrials.gov ID, NCT02978690), and Effisayil™ 1 (NCT03782792), which remains the largest and only randomized clinical trial in patients experiencing GPP flares published to date. In the phase 1 proof-of-concept trial, a Generalized Pustular Psoriasis Physician Global Assessment (GPPGA) score of 0 or 1 (clear or almost clear skin) was attained in 5/7 (71%) patients by week 1 and in all 7 patients by week 4; and the mean percent improvement in the Generalized Pustular Psoriasis Area and Severity Index (GPPASI) score from baseline was 59.0% at week 1, 73.2% at week 2, and 79.8% at week 4. In Effisayil™ 1, a GPPGA pustulation subscore of 0 (no visible pustules) was achieved in 19/35 (54%) patients receiving spesolimab at the end of week 1, versus 1/18 (6%) receiving placebo (difference, 49 percentage points; 95% confidence interval [CI], 21 to 67; P<0.001); and a GPPGA total score of 0 or 1 was achieved by 15/35 (43%) patients in the spesolimab group, versus 2/18 (11%) patients in the placebo group (difference, 32 percentage points; 95% CI, 2 to 53; P = 0.02). Infections at week 1 were reported in 6/35 (17%) patients receiving spesolimab and in 1/18 (6%) patients receiving placebo. These data demonstrate the efficacy and safety of spesolimab in providing rapid and sustained clinical improvement for patients with GPP flares, which translates into improved quality of life, by offering a targeted therapy for GPP.


Sujet(s)
Anticorps monoclonaux humanisés , Agrément de médicaments , Psoriasis , Humains , Psoriasis/traitement médicamenteux , États-Unis , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/effets indésirables , Adulte , Mâle , Femelle , Résultat thérapeutique , Adulte d'âge moyen , Food and Drug Administration (USA) , Interleukines
6.
Cancer Cell ; 42(8): 1333-1335, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39137725

RÉSUMÉ

Glioblastoma is the most common brain cancer, with a 5-year survival rate of less than 10%. This grim prognosis highlights the urgent need for novel therapeutic approaches. In this issue of Cancer Cell, Shanley et al.1 report an innovative engineering strategy to supercharge NK cell immunity against glioblastoma.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Interleukines , Cellules tueuses naturelles , Glioblastome/immunologie , Glioblastome/thérapie , Humains , Cellules tueuses naturelles/immunologie , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/thérapie , Interleukines/métabolisme , Interleukines/immunologie
7.
Cancer Cell ; 42(8): 1450-1466.e11, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39137729

RÉSUMÉ

Glioblastoma (GBM) is an aggressive brain cancer with limited therapeutic options. Natural killer (NK) cells are innate immune cells with strong anti-tumor activity and may offer a promising treatment strategy for GBM. We compared the anti-GBM activity of NK cells engineered to express interleukin (IL)-15 or IL-21. Using multiple in vivo models, IL-21 NK cells were superior to IL-15 NK cells both in terms of safety and long-term anti-tumor activity, with locoregionally administered IL-15 NK cells proving toxic and ineffective at tumor control. IL-21 NK cells displayed a unique chromatin accessibility signature, with CCAAT/enhancer-binding proteins (C/EBP), especially CEBPD, serving as key transcription factors regulating their enhanced function. Deletion of CEBPD resulted in loss of IL-21 NK cell potency while its overexpression increased NK cell long-term cytotoxicity and metabolic fitness. These results suggest that IL-21, through C/EBP transcription factors, drives epigenetic reprogramming of NK cells, enhancing their anti-tumor efficacy against GBM.


Sujet(s)
Tumeurs du cerveau , Protéine delta liant les séquences stimulatrices de type CCAAT , Glioblastome , Interleukines , Cellules tueuses naturelles , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Glioblastome/immunologie , Glioblastome/génétique , Glioblastome/anatomopathologie , Glioblastome/thérapie , Interleukines/génétique , Interleukines/métabolisme , Interleukines/immunologie , Humains , Animaux , Souris , Protéine delta liant les séquences stimulatrices de type CCAAT/métabolisme , Protéine delta liant les séquences stimulatrices de type CCAAT/génétique , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/thérapie , Lignée cellulaire tumorale , Interleukine-15/génétique , Interleukine-15/métabolisme , Interleukine-15/immunologie , Tests d'activité antitumorale sur modèle de xénogreffe
8.
Medicine (Baltimore) ; 103(32): e39036, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39121248

RÉSUMÉ

The association between interleukins and osteoporosis has attracted much attention these days. However, the causal relationship between them is uncertain. Hence, this study performed a Mendelian randomization (MR) analysis to investigate the causal effects of interleukins on osteoporosis. The summary data for interleukins and osteoporosis came from 4 different genome-wide association studies. Significant and independent (P < 5 × 10-6; r2 < 0.001, 10,000 kbp) single-nucleotide polymorphisms were extracted for MR analysis. The inverse-variance weighted and other methods were used for MR analysis, while sensitivity analyses were conducted to test the reliability and stability. The positive causal effects of interleukin-7 on osteoporosis (odds ratio = 1.084; 95% confidence interval: 1.010-1.163; P = .025) were observed. No causal relationship was found between other interleukins and osteoporosis. In the sensitivity analysis, the results did not show the presence of pleiotropy and heterogeneity. Therefore, the results were robust for the MR analysis. This study revealed that interleukin-7 was positively related to osteoporosis and that other interleukins were not related to osteoporosis.


Sujet(s)
Étude d'association pangénomique , Interleukines , Analyse de randomisation mendélienne , Ostéoporose , Polymorphisme de nucléotide simple , Humains , Ostéoporose/génétique , Interleukines/génétique , Interleukine-7/génétique , Prédisposition génétique à une maladie
9.
J Clin Invest ; 134(15)2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-39087473

RÉSUMÉ

Inflammatory neuropathies, which include chronic inflammatory demyelinating polyneuropathy (CIDP) and Guillain Barré syndrome (GBS), result from autoimmune destruction of the PNS and are characterized by progressive weakness and sensory loss. CD4+ T cells play a key role in the autoimmune destruction of the PNS. Yet, key properties of pathogenic CD4+ T cells remain incompletely understood. Here, we used paired single-cell RNA-Seq (scRNA-Seq) and single-cell T cell receptor-sequencing (scTCR-Seq) of peripheral nerves from an inflammatory neuropathy mouse model to identify IL-21-expressing CD4+ T cells that were clonally expanded and multifunctional. These IL-21-expressing CD4+ T cells consisted of 2 transcriptionally distinct expanded cell populations, which expressed genes associated with T follicular helper (Tfh) and T peripheral helper (Tph) cell subsets. Remarkably, TCR clonotypes were shared between these 2 IL-21-expressing cell populations, suggesting a common lineage differentiation pathway. Finally, we demonstrated that IL-21 receptor-KO (IL-21R-KO) mice were protected from neuropathy development and had decreased immune infiltration into peripheral nerves. IL-21 signaling upregulated CXCR6, a chemokine receptor that promotes CD4+ T cell localization in peripheral nerves. Together, these findings point to IL-21 signaling, Tfh/Tph differentiation, and CXCR6-mediated cellular localization as potential therapeutic targets in inflammatory neuropathies.


Sujet(s)
Lymphocytes T CD4+ , Interleukines , Animaux , Souris , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/anatomopathologie , Lignage cellulaire , Interleukines/génétique , Interleukines/immunologie , Interleukines/métabolisme , Souris knockout , Mâle , Femelle
10.
Int J Mol Sci ; 25(15)2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39125732

RÉSUMÉ

Bone metastases, a common and debilitating consequence of advanced cancers, involve a complex interplay between malignant cells and the bone microenvironment. Central to this interaction are interleukins (ILs), a group of cytokines with critical roles in immune modulation and inflammation. This review explores the dualistic nature of pro-inflammatory and anti-inflammatory interleukins in bone metastases, emphasizing their molecular mechanisms, pathological impacts, and therapeutic potential. Pro-inflammatory interleukins, such as IL-1, IL-6, and IL-8, have been identified as key drivers in promoting osteoclastogenesis, tumor proliferation, and angiogenesis. These cytokines create a favorable environment for cancer cell survival and bone degradation, contributing to the progression of metastatic lesions. Conversely, anti-inflammatory interleukins, including IL-4, IL-10, and IL-13, exhibit protective roles by modulating immune responses and inhibiting osteoclast activity. Understanding these opposing effects is crucial for developing targeted therapies aimed at disrupting the pathological processes in bone metastases. Key signaling pathways, including NF-κB, JAK/STAT, and MAPK, mediate the actions of these interleukins, influencing tumor cell survival, immune cell recruitment, and bone remodeling. Targeting these pathways presents promising therapeutic avenues. Current treatment strategies, such as the use of denosumab, tocilizumab, and emerging agents like bimekizumab and ANV419, highlight the potential of interleukin-targeted therapies in mitigating bone metastases. However, challenges such as therapeutic resistance, side effects, and long-term efficacy remain significant hurdles. This review also addresses the potential of interleukins as diagnostic and prognostic biomarkers, offering insights into patient stratification and personalized treatment approaches. Interleukins have multifaceted roles that depend on the context, including the environment, cell types, and cellular interactions. Despite substantial progress, gaps in research persist, particularly regarding the precise mechanisms by which interleukins influence the bone metastatic niche and their broader clinical implications. While not exhaustive, this overview underscores the critical roles of interleukins in bone metastases and highlights the need for continued research to fully elucidate their complex interactions and therapeutic potential. Addressing these gaps will be essential for advancing our understanding and treatment of bone metastases in cancer patients.


Sujet(s)
Tumeurs osseuses , Interleukines , Microenvironnement tumoral , Humains , Tumeurs osseuses/secondaire , Tumeurs osseuses/métabolisme , Interleukines/métabolisme , Animaux , Transduction du signal
11.
Int J Mol Sci ; 25(15)2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39126011

RÉSUMÉ

The clinical manifestations of atopic dermatitis (AD) and chronic nodular prurigo (CNPG) include pruritus and eczema/lesions, posing significant challenges for patients. Th2 cells and ILC2, marked by cytokine production-particularly IL-4/13-are crucial therapeutic targets. Despite displaying a dose-dependent lack of pruritus induction post-injection, IL-13 acts through the IL-13Rα1 and IL-13Rα2 receptor system. Our study focused on investigating ex vivo skin biopsies in AD (n = 17), CNPG (n = 14) and healthy controls (HC; n = 10), examining the gene expression landscape of interleukins linked with pruritus (IL-13, IL-4, IL-31) and their corresponding receptors. Compared to HC, results revealed a significant upregulation of IL-4, IL-13, and IL-13RA1 in AD, whereas CNPG did not show increased IL13 expression. Notably, the decoy receptor IL-13RA2 displayed intriguing patterns, with AD showing a marked increase compared to both HC and CNPG. Positive correlations between receptor expression and itch intensity and hyperkinesis sensation underscore clinical relevance, potentially serving as biomarkers. The findings suggest a pivotal role of IL-4 and IL-13, along with IL-13RA1, in pruritus pathogenesis in both entities, while IL-13 upregulation in AD is countered by IL-13RA2. The comparable expression of IL-13RA2 to HC in CNPG suggests the absence of this regulatory mechanism, potentially worsening the disease and leading to prolonged scratching behavior. These insights illuminate the intricate interplay of interleukins and receptors in different pruritus phenotypes, laying the groundwork for understanding underlying mechanisms and offering avenues for therapeutic intervention.


Sujet(s)
Eczéma atopique , Interleukine-13 , Interleukines , Prurigo , Prurit , Humains , Eczéma atopique/métabolisme , Eczéma atopique/génétique , Eczéma atopique/anatomopathologie , Eczéma atopique/immunologie , Prurigo/métabolisme , Prurigo/anatomopathologie , Prurigo/génétique , Femelle , Adulte , Mâle , Interleukine-13/métabolisme , Interleukine-13/génétique , Interleukines/métabolisme , Interleukines/génétique , Prurit/métabolisme , Prurit/génétique , Adulte d'âge moyen , Interleukine-4/métabolisme , Interleukine-4/génétique , Maladie chronique , Peau/métabolisme , Peau/anatomopathologie , Jeune adulte , Sous-unité alpha1 du récepteur à l'interleukine-13/métabolisme , Sous-unité alpha1 du récepteur à l'interleukine-13/génétique , Sous-unité alpha2 du récepteur à l'interleukine-13/métabolisme , Sous-unité alpha2 du récepteur à l'interleukine-13/génétique
12.
Signal Transduct Target Ther ; 9(1): 199, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39117617

RÉSUMÉ

High frequencies of stem-like memory T cells in infusion products correlate with superior patient outcomes across multiple T cell therapy trials. Herein, we analyzed a published CRISPR activation screening to identify transcriptional regulators that could be harnessed to augment stem-like behavior in CD8+ T cells. Using IFN-γ production as a proxy for CD8+ T cell terminal differentiation, LMO4 emerged among the top hits inhibiting the development of effectors cells. Consistently, we found that Lmo4 was downregulated upon CD8+ T cell activation but maintained under culture conditions facilitating the formation of stem-like T cells. By employing a synthetic biology approach to ectopically express LMO4 in antitumor CD8+ T cells, we enabled selective expansion and enhanced persistence of transduced cells, while limiting their terminal differentiation and senescence. LMO4 overexpression promoted transcriptional programs regulating stemness, increasing the numbers of stem-like CD8+ memory T cells and enhancing their polyfunctionality and recall capacity. When tested in syngeneic and xenograft tumor models, LMO4 overexpression boosted CD8+ T cell antitumor immunity, resulting in enhanced tumor regression. Rather than directly modulating gene transcription, LMO4 bound to JAK1 and potentiated STAT3 signaling in response to IL-21, inducing the expression of target genes (Tcf7, Socs3, Junb, and Zfp36) crucial for memory responses. CRISPR/Cas9-deletion of Stat3 nullified the enhanced memory signature conferred by LMO4, thereby abrogating the therapeutic benefit of LMO4 overexpression. These results establish LMO4 overexpression as an effective strategy to boost CD8+ T cell stemness, providing a new synthetic biology tool to bolster the efficacy of T cell-based immunotherapies.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Lymphocytes T CD8+ , Protéines à domaine LIM , Facteur de transcription STAT-3 , Transduction du signal , Protéines à domaine LIM/génétique , Protéines à domaine LIM/immunologie , Lymphocytes T CD8+/immunologie , Facteur de transcription STAT-3/génétique , Facteur de transcription STAT-3/immunologie , Facteur de transcription STAT-3/métabolisme , Souris , Animaux , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/immunologie , Humains , Transduction du signal/immunologie , Transduction du signal/génétique , Interleukines/génétique , Interleukines/immunologie , Différenciation cellulaire/génétique , Différenciation cellulaire/immunologie
13.
Arch Dermatol Res ; 316(8): 561, 2024 Aug 23.
Article de Anglais | MEDLINE | ID: mdl-39177716

RÉSUMÉ

Circular RNAs (circRNAs) are demonstrated to be involved in psoriasis progression. CircRNAs can act as RNA-binding protein (RBP) sponges. Here, we investigated the action of circAKR1B10 in psoriasis, and explored the potential proteins interacted with circAKR1B10. Levels of genes and proteins were assayed by qRT-PCR and western blotting analyses. Keratinocytes in functional groups were treated with interleukin (IL)-22. Functional analysis were conducted using MTT, 5-ethynyl-2'-deoxyuridine (EdU), and transwell assays, respectively. Interaction analysis among circAKR1B10, Eukaryotic initiation factor 4 A-III (EIF4A3) and Aurora Kinase A (AURKA) was conducted using bioinformatics analysis, RNA pull-down assay, and RNA immunoprecipitation (RIP) assay. CircAKR1B10 was highly expressed in psoriasis patients and IL-22-induced keratinocytes. Functionally, knockdown of circAKR1B10 abolished IL-22-induced proliferation, migration and invasion in keratinocytes. AURKA expression was also higher in psoriasis patients and IL-22-induced keratinocytes, and was negatively correlated with circAKR1B10 expression. Moreover, AURKA silencing reduced the proliferative, migratory and invasive abilities of IL-22-induced keratinocytes. Mechanistically, circAKR1B10 interacted with EIF4A3 protein to stabilize and regulate AURKA expression. CircAKR1B10 contributes to IL-22-induced proliferation, migration and invasion in keratinocytes via up-regulating AURKA expression through interacting with EIF4A3 protein.


Sujet(s)
Aurora kinase A , Mouvement cellulaire , Prolifération cellulaire , Facteur-4A d'initiation eucaryote , , Interleukines , Kératinocytes , Psoriasis , ARN circulaire , Humains , Aurora kinase A/métabolisme , Aurora kinase A/génétique , ARN circulaire/génétique , ARN circulaire/métabolisme , Kératinocytes/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Psoriasis/anatomopathologie , Psoriasis/métabolisme , Psoriasis/génétique , Facteur-4A d'initiation eucaryote/métabolisme , Facteur-4A d'initiation eucaryote/génétique , Interleukines/métabolisme , Interleukines/génétique , DEAD-box RNA helicases
14.
Mol Med Rep ; 30(4)2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39129303

RÉSUMÉ

Diospyros lotus has been traditionally used in Asia for medicinal purposes, exhibiting a broad spectrum of pharmacological effects including antioxidant, neuroprotective and anti­inflammatory properties. While the anti­itch effect of D. lotus leaves has been reported, studies on the detailed mechanism of action in microglia and astrocytes, which are members of the central nervous system, have yet to be revealed. The present study aimed to investigate effects of D. lotus leaf extract (DLE) and its main component myricitrin (MC) on itch­related cytokines and signaling pathways in lipopolysaccharide (LPS)­stimulated microglia. The effect of DLE and MC on activation of astrocyte stimulated by microglia was also examined. Cytokine production was evaluated through reverse transcription PCR and western blot analysis. Signaling pathway was analyzed by performing western blotting and immunofluorescence staining. The effect of microglia on astrocytes activation was evaluated via western blotting for receptors, signaling molecules and itch mediators and confirmed through gene silencing using short interfering RNA. DLE and MC suppressed the production of itch­related cytokine IL­6 and IL­31 in LPS­stimulated microglia. These inhibitory effects were mediated through the blockade of NF­κB, MAPK and JAK/STAT pathways. In astrocytes, stimulation by microglia promoted the expression of itch­related molecules such as oncostatin M receptor, interleukin 31 receptor a, inositol 1,4,5­trisphosphate receptor 1, lipocalin­2 (LCN2), STAT3 and glial fibrillary acidic protein. However, DLE and MC significantly inhibited these receptors. Additionally, astrocytes stimulated by microglia with IL­6, IL­31, or both genes silenced did not show activation of LCN2 or STAT3. The findings of the present study demonstrated that DLE and MC could suppress pruritic activity in astrocytes induced by microglia­derived IL­6 and IL­31. This suggested the potential of DLE and MC as functional materials capable of alleviating pruritus.


Sujet(s)
Astrocytes , Diospyros , Flavonoïdes , Interleukine-6 , Microglie , Extraits de plantes , Feuilles de plante , Prurit , Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , Animaux , Flavonoïdes/pharmacologie , Flavonoïdes/composition chimique , Souris , Interleukine-6/métabolisme , Interleukine-6/génétique , Feuilles de plante/composition chimique , Prurit/traitement médicamenteux , Prurit/métabolisme , Diospyros/composition chimique , Lipopolysaccharides , Transduction du signal/effets des médicaments et des substances chimiques , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Interleukines
15.
Zhongguo Fei Ai Za Zhi ; 27(7): 550-558, 2024 Jul 20.
Article de Chinois | MEDLINE | ID: mdl-39147710

RÉSUMÉ

Non-small cell lung cancer (NSCLC) is a prevalent and aggressive global malignancy. Conventional surgical treatments, radiotherapy, chemotherapy, and targeted therapies often fall short in halting disease progression due to inherent limitations, resulting in suboptimal prognosis. Despite the advent of immunotherapy drugs offering new hope for NSCLC treatment, current efficacy remains insufficient to meet all patient needs. Therefore, actively exploring novel immunotherapeutic approaches to further reduce mortality rates in NSCLC patients has become a crucial focus of NSCLC research. This article aims to systematically review the anti-tumor effects of interleukin-21 and follicular helper T cells in NSCLC immunotherapy by summarizing and analyzing relevant literatures from both domestic and international sources, as well as exploring the potential for enhancing NSCLC treatment prospects through immune checkpoint regulation via immunotherapeutic means.
.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Immunothérapie , Interleukines , Tumeurs du poumon , Humains , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/thérapie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Tumeurs du poumon/immunologie , Tumeurs du poumon/thérapie , Tumeurs du poumon/traitement médicamenteux , Interleukines/immunologie , Interleukines/usage thérapeutique , Immunothérapie/méthodes , Animaux
16.
JCI Insight ; 9(16)2024 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-39171524

RÉSUMÉ

Decidual regulatory T cells (Tregs) are essential for successful pregnancy outcome. A subset of Tregs, T cell immunoglobulin and mucin domain-containing protein 3-positive regulatory T cells (TregsTim-3+), plays a central role in the acceptance of the fetus during early stages of normal pregnancy. The molecular mechanism regulating the differentiation and function of TregsTim-3+ is unknown. Here, we investigated the role of the transcription factor B lymphocyte-induced maturation protein 1 (Blimp-1) on decidual TregTim-3+ differentiation. We demonstrated that Blimp-1 enhanced the coexpression of negative costimulatory molecules (Tim-3, T cell immunoreceptor with Ig and ITIM domains, and programmed cell death protein 1) on Tregs and improved their immunosuppressive functions, including increased IL-10 secretion, suppression of effector T cell proliferation, and promotion of macrophage polarization toward the M2 phenotype. Furthermore, we showed that IL-27 regulated the expression of Tim-3 and Blimp-1 through the STAT1 signaling pathway and that transfer of TregsBlimp-1+ into an abortion-prone mouse model effectively reduced embryo absorption rate. We postulated that abnormalities in the IL-27/Blimp-1 axis might be associated with recurrent pregnancy loss (RPL). These findings provided insights for developing more efficient immunotherapies for women with RPL.


Sujet(s)
Différenciation cellulaire , Récepteur cellulaire-2 du virus de l'hépatite A , Facteur-1 liant le domaine de régulation positive I , Lymphocytes T régulateurs , Femelle , Animaux , Grossesse , Facteur-1 liant le domaine de régulation positive I/métabolisme , Facteur-1 liant le domaine de régulation positive I/génétique , Récepteur cellulaire-2 du virus de l'hépatite A/métabolisme , Souris , Différenciation cellulaire/immunologie , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Facteur de transcription STAT-1/métabolisme , Transduction du signal , Humains , Caduques/immunologie , Caduques/métabolisme , Caduques/cytologie , Interleukine-10/métabolisme , Interleukines/métabolisme , Interleukines/immunologie , Interleukine-27/métabolisme
17.
Zhongguo Dang Dai Er Ke Za Zhi ; 26(8): 879-886, 2024 Aug 15.
Article de Chinois | MEDLINE | ID: mdl-39148395

RÉSUMÉ

Neonatal sepsis is a common and severe infectious disease with a high mortality rate. Its pathogenesis is complex, lacks specific manifestations, and has a low positive culture rate, making early diagnosis and personalized treatment still a challenge for clinicians. Epidemiological studies on twins have shown that genetic factors are associated with neonatal sepsis. Gene polymorphisms are closely related to susceptibility, disease development, and prognosis. This article provides a review of gene polymorphisms related to neonatal sepsis, including interleukins, tumor necrosis factor, Toll-like receptors, NOD-like receptors, CD14, triggering receptor expressed on myeloid cells-1, mannose-binding lectin, and other immune proteins, aiming to promote precision medicine for this disease.


Sujet(s)
Prédisposition génétique à une maladie , Sepsis néonatal , Polymorphisme génétique , Humains , Nouveau-né , Sepsis néonatal/génétique , Interleukines/génétique
18.
Korean J Gastroenterol ; 84(2): 65-81, 2024 Aug 25.
Article de Coréen | MEDLINE | ID: mdl-39176462

RÉSUMÉ

Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is a chronic inflammatory disease of the gastrointestinal tract. The introduction of biologics, particularly anti-interleukin (IL) agents, has revolutionized IBD treatment. This review summarizes the role of ILs in IBD pathophysiology and describes the efficacy and positioning of anti-IL therapies. We discuss the functions of key ILs in IBD and their potential as therapeutic targets. The review then discusses anti-IL therapies, focusing primarily on ustekinumab (anti-IL-12/23), risankizumab (anti-IL-23), and mirikizumab (anti-IL-23). Clinical trial data demonstrate their efficacy in inducing and maintaining remission in Crohn's disease and ulcerative colitis. The safety profiles of these agents are generally favorable. However, long-term safety data for newer agents are still limited. The review also briefly discusses emerging therapies such as guselkumab and brazikumab. Network meta-analyses suggest that anti-IL therapies perform well compared to other biological agents. These agents may be considered first- or second-line therapies for many patients, especially those with comorbidities or safety concerns. Anti-IL therapies represent a significant advancement in IBD treatment, offering effective and relatively safe options for patients with moderate to severe disease.


Sujet(s)
Anticorps monoclonaux , Maladies inflammatoires intestinales , Ustékinumab , Humains , Maladies inflammatoires intestinales/traitement médicamenteux , Ustékinumab/usage thérapeutique , Anticorps monoclonaux/usage thérapeutique , Interleukines/métabolisme , Interleukines/antagonistes et inhibiteurs , Produits biologiques/usage thérapeutique , Anticorps monoclonaux humanisés/usage thérapeutique , Interleukine-23/antagonistes et inhibiteurs , Interleukine-23/métabolisme , Maladie de Crohn/traitement médicamenteux
19.
J Neuroimmune Pharmacol ; 19(1): 37, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39052165

RÉSUMÉ

Recombinant interleukin-22 (rIL-22) has been reported as a protective agent in murine models of diseases driven by epithelial injury. Parasites have a circadian rhythm and their sensitivity to a certain drug may vary during the day. Therefore, this work aimed to investigate the effect of rIL-22 administration at different times of the day on the inflammation, oxidative status, and neurotransmitter release in the gut-brain axis of the Schistosoma mansoni-infected mice. Sixty male BALB/c mice aged six weeks weighing 25-30 g were divided into a control group (injected intraperitoneally with PBS), mice infected with 80 ± 10 cercariae of S. mansoni (infected group) then injected intraperitoneally with PBS, and rIL-22 treated groups. rIL-22 was administrated intraperitoneally (400 ng/kg) either at the onset or offset of the light phase for 14 days. IL-22 administration reduced the levels of IL-1ß, tumor necrosis factor-alpha (TNF-α), nuclear factor kappa beta (NF-κß), and enhanced the production of IL-22 and IL-17. The treatment with IL-22 increased glutathione (GSH) and reduced malondialdehyde (MDA) and nitric oxide (NO) levels both in the ileum and brain. The B-cell lymphoma 2 (BCL2) protein level in the ileum was diminished after IL-22 administration. Brain-derived neurotrophic factor (BDNF) and neurotransmitter release (serotonin, 5HT, norepinephrine, NE, dopamine, DA, Glutamate, Glu, and -amino butyric acid, GABA) were improved by rIL-22. In conclusion, rIL-22 showed promising immunotherapy for inflammation, oxidative damage, and neuropathological signs associated with schistosomiasis. The efficacy of IL-22 increased significantly upon its administration at the time of light offset.


Sujet(s)
Axe cerveau-intestin , , Interleukines , Souris de lignée BALB C , Agents neuromédiateurs , Protéines recombinantes , Schistosomiase à Schistosoma mansoni , Animaux , Souris , Mâle , Agents neuromédiateurs/métabolisme , Agents neuromédiateurs/pharmacologie , Interleukines/métabolisme , Schistosomiase à Schistosoma mansoni/traitement médicamenteux , Schistosomiase à Schistosoma mansoni/immunologie , Schistosomiase à Schistosoma mansoni/métabolisme , Protéines recombinantes/pharmacologie , Protéines recombinantes/administration et posologie , Axe cerveau-intestin/effets des médicaments et des substances chimiques , Axe cerveau-intestin/physiologie , Immunothérapie/méthodes , Monoamines biogènes/métabolisme , Inflammation/métabolisme , Inflammation/traitement médicamenteux
20.
Int J Mol Sci ; 25(14)2024 Jul 21.
Article de Anglais | MEDLINE | ID: mdl-39063193

RÉSUMÉ

Cytokine storm is usually described as one of the main reasons behind COVID-associated mortality. Cytokines are essential protein molecules engaged in immune responses; they play a critical role in protection against infections. However, they also contribute to inflammatory reactions and tissue damage, becoming a double-edged sword in the context of COVID-19. Recent studies have suggested various cytokines and chemokines that play a crucial role in the immune response to SARS-CoV-2 infection. One such cytokine is interleukin 27 (IL-27), which has been found to be elevated in the blood plasma of patients with COVID-19. Within this study, we will explore the role of IL-27 in immune responses and analyze both the existing literature and our own prior research findings on this cytokine in the context of COVID-19. It affects a wide variety of immune cells. Regardless of the pathological process it is involved in, IL-27 is critical for upholding the necessary balance between tissue damage and cytotoxicity against infectious agents and/or tumors. In COVID-19, it is involved in multiple processes, including antiviral cytotoxicity via CD8+ cells, IgG subclass switching, and even the activation of Tregs.


Sujet(s)
COVID-19 , SARS-CoV-2 , COVID-19/immunologie , Humains , SARS-CoV-2/immunologie , Syndrome de libération de cytokines/immunologie , Lymphocytes T CD8+/immunologie , Interleukine-27/métabolisme , Lymphocytes T régulateurs/immunologie , Interleukines/immunologie , Interleukines/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE