Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.470
Filtrer
1.
Int Immunopharmacol ; 136: 112383, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-38843642

RÉSUMÉ

The treatment of autoimmune and inflammatory diseases often requires targeting multiple pathogenic pathways. KYS202004A is a novel bispecific fusion protein designed to antagonize TNF-α and IL-17A, pivotal in the pathophysiology of autoimmune and inflammatory diseases. Our initial efforts focused on screening for optimal structure by analyzing expression levels, purity, and binding capabilities. The binding affinity of KYS202004A to TNF-α and IL-17A was evaluated using SPR. In vitro, we assessed the inhibitory capacity of KYS202004A on cytokine-induced CXCL1 expression in HT29 cells. In vivo, its efficacy was tested using a Collagen-Induced Arthritis (CIA) model in transgenic human-IL-17A mice and an imiquimod-induced psoriasis model in cynomolgus monkeys. KYS202004A demonstrated significant inhibition of IL-17A and TNF-α signaling pathways, outperforming the efficacy of monotherapeutic agents ixekizumab and etanercept in reducing CXCL1 expression in vitro and ameliorating disease markers in vivo. In the CIA model, KYS202004A significantly reduced clinical symptoms, joint destruction, and serum IL-6 concentrations. The psoriasis model revealed that KYS202004A, particularly at a 2  mg/kg dose, was as effective as the combination of ixekizumab and etanercept. This discovery represents a significant advancement in treating autoimmune and inflammatory diseases, offering a dual-targeted therapeutic approach with enhanced efficacy over current monotherapies.


Sujet(s)
Arthrite expérimentale , Interleukine-17 , Macaca fascicularis , Psoriasis , Protéines de fusion recombinantes , Facteur de nécrose tumorale alpha , Animaux , Interleukine-17/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Humains , Psoriasis/traitement médicamenteux , Psoriasis/immunologie , Psoriasis/induit chimiquement , Protéines de fusion recombinantes/usage thérapeutique , Protéines de fusion recombinantes/pharmacologie , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/immunologie , Souris , Chimiokine CXCL1/métabolisme , Chimiokine CXCL1/génétique , Cellules HT29 , Maladies auto-immunes/traitement médicamenteux , Maladies auto-immunes/immunologie , Souris transgéniques , Modèles animaux de maladie humaine , Anticorps bispécifiques/usage thérapeutique , Anticorps bispécifiques/pharmacologie , Mâle , Évaluation préclinique de médicament , Imiquimod , Anti-inflammatoires/usage thérapeutique , Anti-inflammatoires/pharmacologie , Souris de lignée DBA
2.
Int J Mol Sci ; 25(11)2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38891893

RÉSUMÉ

Skin macrophages are critical to maintain and restore skin homeostasis. They serve as major producers of cytokines and chemokines in the skin, participating in diverse biological processes such as wound healing and psoriasis. The heterogeneity and functional diversity of macrophage subpopulations endow them with multifaceted roles in psoriasis development. A distinct subpopulation of skin macrophages, characterized by high expression of CD169, has been reported to exist in both mouse and human skin. However, its role in psoriasis remains unknown. Here, we report that CD169+ macrophages exhibit increased abundance in imiquimod (IMQ) induced psoriasis-like skin lesions. Specific depletion of CD169+ macrophages in CD169-ditheria toxin receptor (CD169-DTR) mice inhibits IMQ-induced psoriasis, resulting in milder symptoms, diminished proinflammatory cytokine levels and reduced proportion of Th17 cells within the skin lesions. Furthermore, transcriptomic analysis uncovers enhanced activity in CD169+ macrophages when compared with CD169- macrophages, characterized by upregulated genes that are associated with cell activation and cell metabolism. Mechanistically, CD169+ macrophages isolated from IMQ-induced skin lesions produce more proinflammatory cytokines and exhibit enhanced ability to promote Th17 cell differentiation in vitro. Collectively, our findings highlight the crucial involvement of CD169+ macrophages in psoriasis development and offer novel insights into the heterogeneity of skin macrophages in the context of psoriasis.


Sujet(s)
Imiquimod , Macrophages , Psoriasis , Lectine-1 de type Ig liant l'acide sialique , Peau , Animaux , Psoriasis/immunologie , Psoriasis/métabolisme , Psoriasis/anatomopathologie , Psoriasis/induit chimiquement , Psoriasis/génétique , Macrophages/métabolisme , Macrophages/immunologie , Souris , Peau/métabolisme , Peau/anatomopathologie , Peau/immunologie , Lectine-1 de type Ig liant l'acide sialique/métabolisme , Cytokines/métabolisme , Modèles animaux de maladie humaine , Cellules Th17/immunologie , Cellules Th17/métabolisme , Différenciation cellulaire , Souris de lignée C57BL
3.
Int J Immunopathol Pharmacol ; 38: 3946320241260262, 2024.
Article de Anglais | MEDLINE | ID: mdl-38876119

RÉSUMÉ

INTRODUCTION: TYK2 inhibitors and traditional natural drugs as promising drugs for psoriasis therapy are receiving increasing attention. They both affect different molecules of JAK/STAT pathway, but it is currently unclear whether their combination will enhance the effect on psoriasis. In this study, we used imiquimod (IMQ)-induced psoriasis mouse model to investigate the therapeutic effects of the combined administration of deucravacitinib (TYK2 inhibitor) and shikonin. METHODS: Aldara cream containing 5% IMQ was used to topically treat the dorsal skin of each mouse for a total of six consecutive days to induce psoriasis. The psoriasis area and severity index (PASI) scores were recorded every day. On the 7th day, skin tissues were taken for histopathological examination and the content of cytokines in skin were evaluated. The frequency of immune cells in peripheral blood, spleen and skin were detected through flow cytometry. RESULTS: Compared to the vehicle control group, the psoriasis symptoms and immune disorder improved significantly in the combination therapy group and deucravacitinib treatment group on the 7th day, and the expressions of p-STAT3 and Ki67 in skin were reduced as well. Moreover, the combined treatment of deucravacitinib and shikonin for psoriasis was superior to the monotherapy group, especially in inhibiting abnormal capillaries proliferation, reducing immune cells infiltration and decreasing the concentration of IL-12p70 in skin. CONCLUSION: The combination of deucravacitinib and shikonin is a promising clinical application.


Sujet(s)
Association de médicaments , Imiquimod , Naphtoquinones , Psoriasis , Peau , Animaux , Psoriasis/traitement médicamenteux , Psoriasis/induit chimiquement , Naphtoquinones/pharmacologie , Naphtoquinones/usage thérapeutique , Souris , Peau/effets des médicaments et des substances chimiques , Peau/anatomopathologie , Peau/métabolisme , Modèles animaux de maladie humaine , Cytokines/métabolisme , Souris de lignée BALB C , Mâle , Femelle , Benzimidazoles , Quinolinone
4.
Molecules ; 29(11)2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38893287

RÉSUMÉ

Psoriasis is a common chronic immune-mediated inflammatory skin disorder. Sophora flavescens Alt. (S. flavescens) has been widely acknowledged in the prevention and treatment of psoriasis. Kushenol F (KSCF) is a natural isopentenyl flavonoid extracted from the root of S. flavescens. We aimed to investigate the effect and mechanism of KSCF on imiquimod (IMQ)-induced psoriasis-like skin lesions in mice. A mouse model of psoriasis was induced with 5% IMQ for 5 days, and the mice were given KSCF dermally for 5 days. Changes in skin morphology, the psoriasis area, the severity index (PASI), and inflammatory factors of psoriasis-like skin lesions were evaluated. Metabolites in the psoriasis-like skin lesions were analyzed with ultra-high-performance liquid chromatography/mass spectrometry followed by a multivariate statistical analysis to identify the differential metabolites and metabolic pathway. The results of the present study confirmed that KSCF significantly reduced PASI scores, epidermal thickening, and epidermal cell proliferation and differentiation. KSCF also reduced the levels of interleukin (IL)-1ß, IL-6, IL-8, IL-17A, IL-22, IL-23, and tumor necrosis factor (TNF)-α in the injured skin tissues while increasing IL-10 content. KSCF significantly regulated metabolites in the skin samples, and a total of 161 significant metabolites were identified. These differential metabolites involved sphingolipid and linoleic acid metabolism and steroid hormone biosynthesis. Collectively, KSCF inhibited the inflammatory response to prevent IMQ-induced psoriasis-like skin lesions in mice by call-backing the levels of 161 endogenous metabolites and affecting their related metabolic pathways. KSCF has the potential to be developed as a topical drug for treating psoriasis symptoms.


Sujet(s)
Modèles animaux de maladie humaine , Imiquimod , Métabolomique , Psoriasis , Psoriasis/induit chimiquement , Psoriasis/traitement médicamenteux , Psoriasis/métabolisme , Psoriasis/anatomopathologie , Animaux , Imiquimod/toxicité , Souris , Chromatographie en phase liquide à haute performance , Métabolomique/méthodes , Métabolome/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Flavonoïdes/pharmacologie , Spectrométrie de masse , Peau/métabolisme , Peau/effets des médicaments et des substances chimiques , Peau/anatomopathologie , Mâle
5.
Theranostics ; 14(8): 3339-3357, 2024.
Article de Anglais | MEDLINE | ID: mdl-38855186

RÉSUMÉ

Rationale: Skin cells actively metabolize nutrients to ensure cell proliferation and differentiation. Psoriasis is an immune-disorder-related skin disease with hyperproliferation in epidermal keratinocytes and is increasingly recognized to be associated with metabolic disturbance. However, the metabolic adaptations and underlying mechanisms of epidermal hyperproliferation in psoriatic skin remain largely unknown. Here, we explored the role of metabolic competition in epidermal cell proliferation and differentiation in psoriatic skin. Methods: Bulk- and single-cell RNA-sequencing, spatial transcriptomics, and glucose uptake experiments were used to analyze the metabolic differences in epidermal cells in psoriasis. Functional validation in vivo and in vitro was done using imiquimod-like mouse models and inflammatory organoid models. Results: We observed the highly proliferative basal cells in psoriasis act as the winners of the metabolic competition to uptake glucose from suprabasal cells. Using single-cell metabolic analysis, we found that the "winner cells" promote OXPHOS pathway upregulation by COX7B and lead to increased ROS through glucose metabolism, thereby promoting the hyperproliferation of basal cells in psoriasis. Also, to prevent toxic damage from ROS, basal cells activate the glutathione metabolic pathway to increase their antioxidant capacity to assist in psoriasis progression. We further found that COX7B promotes psoriasis development by modulating the activity of the PPAR signaling pathway by bulk RNA-seq analysis. We also observed glucose starvation and high expression of SLC7A11 that causes suprabasal cell disulfide stress and affects the actin cytoskeleton, leading to immature differentiation of suprabasal cells in psoriatic skin. Conclusion: Our study demonstrates the essential role of cellular metabolic competition for skin tissue homeostasis.


Sujet(s)
Différenciation cellulaire , Prolifération cellulaire , Glucose , Kératinocytes , Psoriasis , Psoriasis/métabolisme , Psoriasis/anatomopathologie , Glucose/métabolisme , Humains , Animaux , Souris , Kératinocytes/métabolisme , Modèles animaux de maladie humaine , Analyse sur cellule unique , Cellules épidermiques/métabolisme , Espèces réactives de l'oxygène/métabolisme , Métabolisme énergétique , Épiderme/métabolisme , Épiderme/anatomopathologie , Imiquimod , Mâle
6.
Front Immunol ; 15: 1398120, 2024.
Article de Anglais | MEDLINE | ID: mdl-38903528

RÉSUMÉ

Psoriasis is an inflammatory disease with systemic manifestations that most commonly presents as itchy, erythematous, scaly plaques on extensor surfaces. Activation of the IL-23/IL-17 pro-inflammatory signaling pathway is a hallmark of psoriasis and its inhibition is key to clinical management. Granzyme K (GzmK) is an immune cell-secreted serine protease elevated in inflammatory and proliferative skin conditions. In the present study, human psoriasis lesions exhibited elevated GzmK levels compared to non-lesional psoriasis and healthy control skin. In an established murine model of imiquimod (IMQ)-induced psoriasis, genetic loss of GzmK significantly reduced disease severity, as determined by delayed plaque formation, decreased erythema and desquamation, reduced epidermal thickness, and inflammatory infiltrate. Molecular characterization in vitro revealed that GzmK contributed to macrophage secretion of IL-23 as well as PAR-1-dependent keratinocyte proliferation. These findings demonstrate that GzmK enhances IL-23-driven inflammation as well as keratinocyte proliferation to exacerbate psoriasis severity.


Sujet(s)
Prolifération cellulaire , Granzymes , Inflammation , Interleukine-23 , Kératinocytes , Psoriasis , Psoriasis/immunologie , Psoriasis/anatomopathologie , Animaux , Kératinocytes/métabolisme , Kératinocytes/immunologie , Kératinocytes/anatomopathologie , Humains , Souris , Granzymes/métabolisme , Granzymes/génétique , Interleukine-23/métabolisme , Inflammation/immunologie , Inflammation/anatomopathologie , Imiquimod , Modèles animaux de maladie humaine , Souris knockout , Femelle , Mâle , Souris de lignée C57BL
7.
Nat Commun ; 15(1): 5288, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38902277

RÉSUMÉ

Psoriasis is an immune-mediated skin disease associated with neurogenic inflammation, but the underlying molecular mechanism remains unclear. We demonstrate here that acid-sensing ion channel 3 (ASIC3) exacerbates psoriatic inflammation through a sensory neurogenic pathway. Global or nociceptor-specific Asic3 knockout (KO) in female mice alleviates imiquimod-induced psoriatic acanthosis and type 17 inflammation to the same extent as nociceptor ablation. However, ASIC3 is dispensable for IL-23-induced psoriatic inflammation that bypasses the need for nociceptors. Mechanistically, ASIC3 activation induces the activity-dependent release of calcitonin gene-related peptide (CGRP) from sensory neurons to promote neurogenic inflammation. Botulinum neurotoxin A and CGRP antagonists prevent sensory neuron-mediated exacerbation of psoriatic inflammation to similar extents as Asic3 KO. In contrast, replenishing CGRP in the skin of Asic3 KO mice restores the inflammatory response. These findings establish sensory ASIC3 as a critical constituent in psoriatic inflammation, and a promising target for neurogenic inflammation management.


Sujet(s)
Canaux ioniques sensibles à l'acidité , Peptide relié au gène de la calcitonine , Souris knockout , Psoriasis , Cellules réceptrices sensorielles , Animaux , Canaux ioniques sensibles à l'acidité/métabolisme , Canaux ioniques sensibles à l'acidité/génétique , Femelle , Psoriasis/métabolisme , Psoriasis/anatomopathologie , Psoriasis/génétique , Psoriasis/induit chimiquement , Souris , Peptide relié au gène de la calcitonine/métabolisme , Peptide relié au gène de la calcitonine/génétique , Cellules réceptrices sensorielles/métabolisme , Peau/métabolisme , Peau/anatomopathologie , Imiquimod , Souris de lignée C57BL , Modèles animaux de maladie humaine , Inflammation/métabolisme , Inflammation neurogénique/métabolisme , Humains , Nocicepteurs/métabolisme , Interleukine-23/métabolisme , Interleukine-23/génétique
8.
Ann Clin Microbiol Antimicrob ; 23(1): 57, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38902740

RÉSUMÉ

Chromoblastomycosis (CBM), a chronic fungal infection affecting the skin and subcutaneous tissues, is predominantly caused by dematiaceous fungi in tropical and subtropical areas. Characteristically, CBM presents as plaques and nodules, often leading to scarring post-healing. Besides traditional diagnostic methods such as fungal microscopy, culture, and histopathology, dermatoscopy and reflectance confocal microscopy can aid in diagnosis. The treatment of CBM is an extended and protracted process. Imiquimod, acting as an immune response modifier, boosts the host's immune response against CBM, and controls scar hyperplasia, thereby reducing the treatment duration. We present a case of CBM in Guangdong with characteristic reflectance confocal microscopy manifestations, effectively managed through a combination of itraconazole, terbinafine, and imiquimod, shedding light on novel strategies for managing this challenging condition.


Sujet(s)
Antifongiques , Chromoblastomycose , Imiquimod , Itraconazole , Terbinafine , Chromoblastomycose/traitement médicamenteux , Chromoblastomycose/microbiologie , Imiquimod/usage thérapeutique , Humains , Antifongiques/usage thérapeutique , Itraconazole/usage thérapeutique , Terbinafine/usage thérapeutique , Mâle , Résultat thérapeutique , Microscopie confocale , Peau/anatomopathologie , Peau/microbiologie , Adulte d'âge moyen
9.
Phytomedicine ; 131: 155783, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38838402

RÉSUMÉ

BACKGROUND: Psoriasis, a chronic immune-mediated skin disease with pathological features such as aberrant differentiation of keratinocytes, dermal-epidermal inflammation, and angiogenesis. 2,3,5,4'-Tetrahydroxy stilbene 2-Ο-ß-d-glucoside (2354Glu) is a natural small molecule polyhydrostilbenes isolated from Polygonum multiglorum Thunb. The regulation of IL-36 subfamily has led to new pharmacologic strategies to reverse psoriasiform dermatitis. PURPOSE: Here we investigated the therapeutic potential of 2354Glu and elucidated the underlying mechanism in psoriasis. METHODS: The effects of 2354Glu on IL-36 signaling were assessed by psoriasiform in vivo, in vitro and ex vivo model. The in vivo mice model of psoriasis-like skin inflammation was established by applying imiquimod (IMQ), and the in vitro and ex vitro models were established by stimulating mouse primary keratinocyte, human keratinocytes cells (HaCaT) and ex vivo skin tissue isolated from the mice back with Polyinosine-polycytidylic acid (Poly(I:C)), IMQ, IL-36γ and Lipopolysaccharide (LPS) respectively. Moreover, NETs formation was inhibited by Cl-amidine to evaluate the effect of NETs in psoriatic mouse model. The effects of 2354Glu on skin inflammation were assessed by western blot, H&E, immunohistochemistry, immunofluorescence, enzyme-linked immunosorbent assay and real-time quantitative PCR. RESULTS: In Poly(I:C)-stimulated keratinocytes, the secretion of IL-36 was inhibited after treatment with 2354Glu, similar to the effects of TLR3, P2X7R and caspase-1 inhibitors. In aldara (imiquimod)-induced mice, 2354Glu (100 and 25 mg/kg) improved immune cell infiltration and hyperkeratosis in psoriasis by directly targeting IL-36 in keratinocytes through P2X7R-caspase-1. When treatment with 2354Glu (25 mg/kg) was insufficient to inhibit IL-36γ, NETs reduced pathological features and IL-36 signaling by interacting with keratinocytes to combat psoriasis like inflammation. CONCLUSION: These results indicated that NETs had a beneficial effect on psoriasiform dermatitis. 2354Glu alleviates psoriasis by directly targeting IL-36/P2X7R axis and NET formation, providing a potential candidate for the treatment of psoriasis.


Sujet(s)
Modèles animaux de maladie humaine , Glucosides , Imiquimod , Interleukine-1 , Psoriasis , Stilbènes , Animaux , Psoriasis/traitement médicamenteux , Glucosides/pharmacologie , Humains , Interleukine-1/métabolisme , Stilbènes/pharmacologie , Souris , Kératinocytes/effets des médicaments et des substances chimiques , Polygonum/composition chimique , Peau/effets des médicaments et des substances chimiques , Peau/anatomopathologie , Souris de lignée BALB C , Transduction du signal/effets des médicaments et des substances chimiques , Mâle , Caspase-1/métabolisme
10.
Pharm Res ; 41(6): 1163-1181, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38839718

RÉSUMÉ

OBJECTIVE: This study aims to utilize PEGylated poly (lactic-co-glycolic acid) (PLGA) nanoparticles as a delivery system for simultaneous administration of the BRAFV600E peptide, a tumor-specific antigen, and imiquimod (IMQ). The objective is to stimulate dendritic cell (DC) maturation, activate macrophages, and facilitate antigen presentation in C57BL6 mice. METHODS: PEG-PLGA-IMQ-BRAFV600E nanoparticles were synthesized using a PLGA-PEG-PLGA tri-block copolymer, BRAFV600E, and IMQ. Characterization included size measurement and drug release profiling. Efficacy was assessed in inhibiting BPD6 melanoma cell growth and activating immature bone marrow DCs, T cells, macrophages, and splenocyte cells through MTT and ELISA assays. In vivo, therapeutic and immunogenic effects potential was evaluated, comparing it to IMQ + BRAFV600E and PLGA-IMQ-BRAFV600E nanoparticles in inhibiting subcutaneous BPD6 tumor growth. RESULTS: The results highlight the successful synthesis of PEG-PLGA-IMQ-BRAFV600E nanoparticles (203 ± 11.1 nm), releasing 73.4% and 63.2% of IMQ and BARFV600E, respectively, within the initial 48 h. In vitro, these nanoparticles demonstrated a 1.3-fold increase in potency against BPD6 cells, achieving ~ 2.8-fold enhanced cytotoxicity compared to PLGA-IMQ-BRAFV600E. Moreover, PEG-PLGA-IMQ-BRAFV600E exhibited a 1.3-fold increase in potency for enhancing IMQ cytotoxic effects and a 1.1- to ~ 2.4-fold increase in activating DCs, T cells, macrophages, and splenocyte cells compared to IMQ-BRAFV600E and PLGA-IMQ-BRAFV600E. In vivo, PEG-PLGA-IMQ-BRAFV600E displayed a 1.3- to 7.5-fold increase in potency for inhibiting subcutaneous BPD6 tumor growth compared to the other formulations. CONCLUSIONS: The findings suggest that PEG-PLGA nanoparticles effectively promote DC maturation, T cell activation, and potentially macrophage activation. The study highlights the promising role of this nanocomposite in vaccine development.


Sujet(s)
Cellules dendritiques , Imiquimod , Mélanome , Souris de lignée C57BL , Nanoparticules , Polyéthylène glycols , Protéines proto-oncogènes B-raf , Animaux , Cellules dendritiques/immunologie , Cellules dendritiques/effets des médicaments et des substances chimiques , Polyéthylène glycols/composition chimique , Protéines proto-oncogènes B-raf/génétique , Mélanome/immunologie , Mélanome/traitement médicamenteux , Nanoparticules/composition chimique , Lignée cellulaire tumorale , Souris , Imiquimod/pharmacologie , Adjuvants immunologiques/administration et posologie , Adjuvants immunologiques/pharmacologie , Femelle , Copolymère d'acide poly(lactique-co-glycolique)/composition chimique , Macrophages/effets des médicaments et des substances chimiques , Macrophages/immunologie , Libération de médicament , Humains , Tumeurs cutanées/immunologie , Tumeurs cutanées/prévention et contrôle , Tumeurs cutanées/traitement médicamenteux
11.
J Exp Med ; 221(8)2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-38861030

RÉSUMÉ

Germline gain-of-function (GOF) variants in STAT3 cause an inborn error of immunity associated with early-onset poly-autoimmunity and immune dysregulation. To study tissue-specific immune dysregulation, we used a mouse model carrying a missense variant (p.G421R) that causes human disease. We observed spontaneous and imiquimod (IMQ)-induced skin inflammation associated with cell-intrinsic local Th17 responses in STAT3 GOF mice. CD4+ T cells were sufficient to drive skin inflammation and showed increased Il22 expression in expanded clones. Certain aspects of disease, including increased epidermal thickness, also required the presence of STAT3 GOF in epithelial cells. Treatment with a JAK inhibitor improved skin disease without affecting local Th17 recruitment and cytokine production. These findings collectively support the involvement of Th17 responses in the development of organ-specific immune dysregulation in STAT3 GOF and suggest that the presence of STAT3 GOF in tissues is important for disease and can be targeted with JAK inhibition.


Sujet(s)
Mutation gain de fonction , Imiquimod , Facteur de transcription STAT-3 , Cellules Th17 , Animaux , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique , Cellules Th17/immunologie , Souris , Humains , Imiquimod/pharmacologie , Peau/anatomopathologie , Peau/métabolisme , Peau/immunologie , , Dermatite/immunologie , Dermatite/génétique , Dermatite/anatomopathologie , Dermatite/métabolisme , Souris de lignée C57BL , Interleukines/génétique , Interleukines/métabolisme , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Inflammation/génétique , Inflammation/métabolisme , Inflammation/immunologie , Inflammation/anatomopathologie
12.
Cell Death Dis ; 15(6): 449, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38926337

RÉSUMÉ

Psoriasis is an IL-23/Th17-mediated skin disorder with a strong genetic predisposition. The impact of its susceptibility gene nitric oxide synthase 2 (NOS2) remains unknown. Here, we demonstrate strong NOS2 mRNA expression in psoriatic epidermis, an effect that is IL-17 dependent. However, its complete translation to protein is prevented by the IL-17-induced miR-31 implying marginally upregulated NO levels in psoriatic skin. We demonstrate that lower levels of NO, as opposed to higher levels, increase keratinocyte proliferation and mediate IL-17 downstream effects. We hypothesized that the psoriatic phenotype may be alleviated by either eliminating or increasing cellular NO levels. In fact, using the imiquimod psoriasis mouse model, we found a profound impact on the psoriatic inflammation in both IMQ-treated NOS2 KO mice and wild-type mice treated with IMQ and the NO-releasing berdazimer gel. In conclusion, we demonstrate that IL-17 induces NOS2 and fine-tunes its translation towards a window of proinflammatory and hyperproliferative effects and identify NO donor therapy as a new treatment modality for psoriasis.


Sujet(s)
Interleukine-17 , Souris knockout , Nitric oxide synthase type II , Monoxyde d'azote , Psoriasis , Psoriasis/génétique , Psoriasis/anatomopathologie , Animaux , Nitric oxide synthase type II/métabolisme , Nitric oxide synthase type II/génétique , Souris , Humains , Monoxyde d'azote/métabolisme , Interleukine-17/métabolisme , Kératinocytes/métabolisme , Kératinocytes/anatomopathologie , Kératinocytes/effets des médicaments et des substances chimiques , Imiquimod , Souris de lignée C57BL , Modèles animaux de maladie humaine , Prolifération cellulaire/effets des médicaments et des substances chimiques
14.
Int Immunopharmacol ; 134: 112248, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38749332

RÉSUMÉ

Psoriasis, characterized by aberrant epidermal keratinocyte proliferation and differentiation, is a chronic inflammatory immune-related skin disease. Diosmetin (Dios), derived from citrus fruits, exhibits anti-inflammatory and anti-proliferative properties. In this study, IL-17A-induced HaCaT cell model and Imiquimod (IMQ)-induced mouse model were utilized to investigate the effects of Dios against psoriasis. The morphology and biomarkers of psoriasis were regarded as the preliminary evaluation including PASI score, skin thickness, H&E staining, EdU staining and inflammatory factors. Transcriptomics analysis revealed PGC-1α as a key target for Dios in ameliorating psoriasis. Specifically, Dios, through PGC-1α, suppressed YAP-mediated proliferation and inflammatory responses in psoriatic keratinocytes. In conclusion, Dios shows promise in psoriasis treatment and holds potential for development as targeted medications for application in psoriasis.


Sujet(s)
Prolifération cellulaire , Imiquimod , Kératinocytes , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes , Psoriasis , Transduction du signal , Psoriasis/traitement médicamenteux , Psoriasis/immunologie , Animaux , Kératinocytes/effets des médicaments et des substances chimiques , Kératinocytes/métabolisme , Humains , Transduction du signal/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/génétique , Souris , Flavonoïdes/pharmacologie , Flavonoïdes/usage thérapeutique , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Protéines de signalisation YAP/métabolisme , Modèles animaux de maladie humaine , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Cellules HaCaT , Lignée cellulaire , Souris de lignée BALB C , Interleukine-17/métabolisme , Mâle , Inflammation/traitement médicamenteux
15.
Int Immunopharmacol ; 134: 112261, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38761783

RÉSUMÉ

BACKGROUND: Psoriasis, a chronic inflammatory condition of the skin, is characterized by an atypical proliferation of epidermal keratinocytes and immune cell infiltration. Orientin is a flavonoid monomer with potent anti-inflammatory activities. However, the therapeutic effects of orientin on psoriasis and the underlying mechanisms have not been elucidated. OBJECTIVE: To investigate the therapeutic effect of orientin on psoriasis and the underlying mechanisms using network pharmacology and experimental studies. METHODS: A psoriasis-like mouse model was established using imiquimod (IMQ). Lipopolysaccharide (LPS) was used to stimulate the RAW264.7 and HaCaT cells in vitro. The therapeutic effects of orientin and the underlying mechanism were analyzed using histopathological, immunohistochemical, quantitative real-time polymerase chain reaction, enzyme-linked immunosorbent assay, flow cytometry, and western blotting analyses. RESULTS: Orientin ameliorated skin lesions and suppressed keratinocyte proliferation and immune cell infiltration in the IMQ-induced psoriasis-like mouse model. Additionally, orientin inhibited the secretion of the pro-inflammatory factors interleukin (IL)-1ß, tumor necrosis factor (TNF)-α, IL-6, IL-8, IL-17, and IL-23 in the psoriasis-like mouse model and LPS-induced RAW264.7 and HaCaT cells. Furthermore, orientin mitigated the LPS-induced upregulation of reactive oxygen species and downregulation of IL-10 and glutathione levels. Orientin alleviated inflammation by downregulating the MAPK signaling pathway. CONCLUSION: Orientin alleviated psoriasis-like dermatitis by suppressing the MAPK signaling pathway, suggesting that orientin is a potential therapeutic for psoriasis.


Sujet(s)
Anti-inflammatoires , Cytokines , Modèles animaux de maladie humaine , Flavonoïdes , Glucosides , Cellules HaCaT , Imiquimod , Kératinocytes , Lipopolysaccharides , Système de signalisation des MAP kinases , Souris de lignée BALB C , Psoriasis , Animaux , Psoriasis/traitement médicamenteux , Psoriasis/immunologie , Psoriasis/induit chimiquement , Psoriasis/anatomopathologie , Souris , Humains , Cellules RAW 264.7 , Anti-inflammatoires/usage thérapeutique , Anti-inflammatoires/pharmacologie , Flavonoïdes/pharmacologie , Flavonoïdes/usage thérapeutique , Cytokines/métabolisme , Kératinocytes/effets des médicaments et des substances chimiques , Glucosides/usage thérapeutique , Glucosides/pharmacologie , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Peau/anatomopathologie , Peau/effets des médicaments et des substances chimiques , Peau/immunologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mâle , Espèces réactives de l'oxygène/métabolisme , Dermatite/traitement médicamenteux , Dermatite/anatomopathologie , Dermatite/immunologie , Lignée cellulaire
16.
Phytomedicine ; 130: 155544, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-38810554

RÉSUMÉ

BACKGROUND: Psoriasis is a chronic immune-mediated inflammatory skin disease that affects the quality of life and mental health of approximately 150 million people worldwide. Ze-Qi-Tang (ZQT) is a classic compound used in China for lung disease; however, its mechanism of action in psoriasis remains unclear. This study aimed to investigate the therapeutic effect of the ZQT formula on psoriasis and explore the underlying molecular mechanisms. METHODS: Peripheral blood samples were collected from patients with psoriasis and healthy individuals. Flow cytometry was used to detect changes in the proportions of myeloid-derived suppressor cells (MDSCs) and other immune cells. Psoriasis was induced in mice by the daily application of imiquimod. ZQT was administered separately or in combination with anti-Gr1 antibody to deplete MDSC. The glycolysis levels of the MDSCs were detected using a Seahorse analyzer. The p21/Hif1α/Glut1 pathway was identified and validated by mRNA sequence, RT-qPCR, WB, IF, and the application of p21 inhibitor UC2288. RESULTS: The number of MDSCs was significantly increased in patients with psoriasis, with the increased expression of p21, Hif1α, and Glut1 in MDSCs. ZQT significantly alleviated psoriasis-like skin lesions in mice. ZQT formula significantly reduced the number of MDSCs in psoriatic-like mice and enhanced their suppressive capacity for T cells. The efficacy of ZQT in alleviating psoriatic dermatitis is compromised by MDSC depletion. ZQT decreased the expressions of p21, Hif1α, and Glut1-induced glycolysis in MDSCs, thereby inhibiting Th17 cell differentiation. CONCLUSION: These suggest that ZQT alleviates IMQ-induced psoriatic dermatitis, by inhibiting p21/Hif1α/Glut1-induced glycolysis in MDSCs.


Sujet(s)
Régulation négative , Médicaments issus de plantes chinoises , Transporteur de glucose de type 1 , Glycolyse , Sous-unité alpha du facteur-1 induit par l'hypoxie , Cellules myéloïdes suppressives , Psoriasis , Animaux , Psoriasis/traitement médicamenteux , Transporteur de glucose de type 1/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Glycolyse/effets des médicaments et des substances chimiques , Médicaments issus de plantes chinoises/pharmacologie , Humains , Souris , Cellules myéloïdes suppressives/effets des médicaments et des substances chimiques , Cellules myéloïdes suppressives/métabolisme , Mâle , Régulation négative/effets des médicaments et des substances chimiques , Femelle , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Modèles animaux de maladie humaine , Adulte , Transduction du signal/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Imiquimod , Adulte d'âge moyen
17.
Dev Comp Immunol ; 157: 105197, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38763479

RÉSUMÉ

Toll-like receptors (TLRs) are pivotal pattern recognition receptors (PRRs) and key mediators of innate immunity. Despite the significance of channel catfish (Ictalurus punctatus) in comparative immunology and aquaculture, its 20 TLR genes remain largely functionally uncharacterized. In this study, our aim was to determine the catfish TLR7 agonists, signaling potential, and cellular localization. Using a mammalian reporter system, we identified imiquimod and resiquimod, typical ssRNA analogs, as potent catfish TLR7 agonists. Notably, unlike grass carp TLR7, catfish TLR7 lacks the ability to respond to poly (I:C). Confocal microscopy revealed predominant catfish TLR7 expression in lysosomes, co-localizing with the endosomal chaperone protein, UNC93B1. Furthermore, imiquimod stimulation elicited robust IFNb transcription in peripheral blood leukocytes isolated from adult catfish. These findings underscore the conservation of TLR7 signaling in catfish, reminiscent of mammalian TLR7 responses. Our study sheds light on the functional aspects of catfish TLR7 and contributes to a better understanding of its role in immune defense mechanisms.


Sujet(s)
Protéines de poisson , Ictaluridae , Imidazoles , Imiquimod , Immunité innée , Lysosomes , Récepteur de type Toll-7 , Animaux , Récepteur de type Toll-7/métabolisme , Récepteur de type Toll-7/agonistes , Récepteur de type Toll-7/génétique , Imidazoles/pharmacologie , Ictaluridae/immunologie , Lysosomes/métabolisme , Protéines de poisson/génétique , Protéines de poisson/métabolisme , Transduction du signal , Humains , Aminoquinoléines/pharmacologie , Poly I-C/immunologie
18.
Mol Pharm ; 21(6): 2813-2827, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38752564

RÉSUMÉ

Psoriasis, affecting 2-3% of the global population, is a chronic inflammatory skin condition without a definitive cure. Current treatments focus on managing symptoms. Recognizing the need for innovative drug delivery methods to enhance patient adherence, this study explores a new approach using calcipotriol monohydrate (CPM), a primary topical treatment for psoriasis. Despite its effectiveness, CPM's therapeutic potential is often limited by factors like the greasiness of topical applications, poor skin permeability, low skin retention, and lack of controlled delivery. To overcome these challenges, the study introduces CPM in the form of nanosuspensions (NSs), characterized by an average particle size of 211 ± 2 nm. These CPM NSs are then incorporated into a trilayer dissolving microneedle patch (MAP) made from poly(vinylpyrrolidone) and w poly(vinyl alcohol) as needle arrays and prefrom 3D printed polylactic acid backing layer. This MAP features rapidly dissolving tips and exhibits good mechanical properties and insertion capability with delivery efficiency compared to the conventional Daivonex ointment. The effectiveness of this novel MAP was tested on Sprague-Dawley rats with imiquimod-induced psoriasis, demonstrating efficacy comparable to the marketed ointment. This innovative trilayer dissolving MAP represents a promising new local delivery system for calcipotriol, potentially revolutionizing psoriasis treatment by enhancing drug delivery and patient compliance.


Sujet(s)
Administration par voie cutanée , Calcitriol , Systèmes de délivrance de médicaments , Aiguilles , Psoriasis , Rat Sprague-Dawley , Psoriasis/traitement médicamenteux , Animaux , Calcitriol/analogues et dérivés , Calcitriol/administration et posologie , Rats , Systèmes de délivrance de médicaments/méthodes , Absorption cutanée/effets des médicaments et des substances chimiques , Peau/métabolisme , Peau/effets des médicaments et des substances chimiques , Peau/anatomopathologie , Taille de particule , Mâle , Nanoparticules/composition chimique , Imiquimod/administration et posologie , Suspensions , Produits dermatologiques/administration et posologie , Produits dermatologiques/pharmacocinétique , Patch transdermique
19.
Arch Dermatol Res ; 316(5): 176, 2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38758283

RÉSUMÉ

Psoriasis is a chronic immune mediated inflammatory skin disease with systemic manifestations. It has been reported that caloric restriction could improve severity of psoriasis patients. However, the mechanism of intermittent fasting effects on psoriasis has not been investigated. Caloric restriction is known to reduce the number of circulating inflammatory monocytes in a CCL2-dependent manner. However, it is still unknown whether caloric restriction can improve psoriasis by regulating monocytes through CCL2. In this study, we used imiquimod (IMQ)-induced psoriasis-like mouse model to explore the effects and the mechanisms of intermittent fasting on psoriasis-like dermatitis. We found that intermittent fasting could significantly improve IMQ-induced psoriasis-like dermatitis, and reduce the number of γδT17 cells and IL-17 production in draining lymph nodes and psoriatic lesion via inhibiting proliferation and increasing death of γδT17 cells. Furthermore, intermittent fasting could significantly decrease monocytes in blood, and this was associated with decreased monocytes, macrophages and DC in psoriasis-like skin inflammation. Reduced monocytes in circulation and increased monocytes in BM of fasting IMQ-induced psoriasis-like mice is through reducing the production of CCL2 from BM to inhibit monocyte egress to the periphery. Our above data shads light on the mechanisms of intermittent fasting on psoriasis.


Sujet(s)
Chimiokine CCL2 , Modèles animaux de maladie humaine , Jeûne , Imiquimod , Monocytes , Psoriasis , Animaux , Psoriasis/immunologie , Psoriasis/induit chimiquement , Psoriasis/anatomopathologie , Monocytes/immunologie , Monocytes/métabolisme , Souris , Jeûne/sang , Chimiokine CCL2/métabolisme , Cellules Th17/immunologie , Interleukine-17/métabolisme , Peau/anatomopathologie , Peau/immunologie , Humains , Souris de lignée C57BL , Mâle , Prolifération cellulaire , Restriction calorique , Jeûne intermittent
20.
Int J Nanomedicine ; 19: 4719-4733, 2024.
Article de Anglais | MEDLINE | ID: mdl-38813391

RÉSUMÉ

Introduction: Lung cancer's high incidence and dismal prognosis with traditional treatments like surgery and radiotherapy necessitate innovative approaches. Despite advancements in nanotherapy, the limitations of single-treatment modalities and significant side effects persist. To tackle lung cancer effectively, we devised a temperature-sensitive hydrogel-based local injection system with near-infrared triggered drug release. Utilizing 2D MXene nanosheets as carriers loaded with R837 and cisplatin (DDP), encapsulated within a temperature-sensitive hydrogel-forming PEG-MXene@DDP@R837@SHDS (MDR@SHDS), we administered in situ injections of MDR@SHDS into tumor tissues combined with photothermal therapy (PTT). The immune adjuvant R837 enhances dendritic cell (DC) maturation and tumor cell phagocytosis, while PTT induces tumor cell apoptosis and necrosis by converting light energy into heat energy. Methods: Material characterization employed transmission electron microscopy, X-ray photoelectron spectroscopy, phase transition temperature, and near-infrared thermography. In vitro experiments assessed Lewis cell proliferation and apoptosis using CCK-8, Edu, and TUNEL assays. In vivo experiments on C57 mouse Lewis transplant tumors evaluated the photothermal effect via near-infrared thermography and assessed DC maturation and CD4+/CD8+ T cell ratios using flow cytometry. The in vivo anti-tumor efficacy of MDR@SHDS was confirmed by tumor growth curve recording and HE and TUNEL staining of tumor sections. Results: The hydrogel exhibited excellent temperature sensitivity, controlled release properties, and high biocompatibility. In vitro experiments revealed that MDR@SHDS combined with PTT had a greater inhibitory effect on tumor cell proliferation compared to MDR@SHD alone. Combining local immunotherapy, chemotherapy, and PTT yielded superior anti-tumor effects than individual treatments. Conclusion: MDR@SHDS, with its simplicity, biocompatibility, and enhanced anti-tumor effects in combination with PTT, presents a promising therapeutic approach for lung cancer treatment, offering potential clinical utility.


Sujet(s)
Cisplatine , Imiquimod , Tumeurs du poumon , Souris de lignée C57BL , Animaux , Cisplatine/pharmacologie , Cisplatine/composition chimique , Cisplatine/administration et posologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Souris , Imiquimod/composition chimique , Imiquimod/administration et posologie , Imiquimod/pharmacologie , Hydrogels/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Nanostructures/composition chimique , Thérapie photothermique/méthodes , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/administration et posologie , Lignée cellulaire tumorale , Systèmes de délivrance de médicaments/méthodes , Humains , Température , Cellules dendritiques/effets des médicaments et des substances chimiques , Vecteurs de médicaments/composition chimique , Carcinome pulmonaire de Lewis/traitement médicamenteux , Carcinome pulmonaire de Lewis/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...