Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 73.662
Filtrer
1.
Nutrients ; 16(12)2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38931322

RÉSUMÉ

The immune system is affected by the dietary products humans intake. Immune system regulation by nutrition has uses in the clinical context, but it can also benefit healthy populations by delaying or preventing the emergence of immune-mediated chronic illnesses. In this study, the purpose was to describe and compare the modulator effects on the immune system of the routine ingestion of fresh vs. pasteurized yogurt. A unicentral, prospective, randomized, double-blind, parallel group 8-week nutritional study was carried out comparing the ingestion of 125 g of the products in healthy adults three times a day. A complete battery of in vitro tests on the activity of the immune system, processes and phenomena was performed. Exclusive immune-modulatory effects of fresh yogurt with respect to base line were found in terms of increased systemic IgM (primary immune responses), increased synthesis of IFN-gamma upon stimulation (Th1) and increased peripheral T cells (mainly "naive" CD4s). In the three interventions, we observed an increased phagocytic activity and burst test in granulocytes, together with increased secretion of IL-6, IL-1 ß and IL-8 (pro-inflammatory) and increased CD16 expression (FcR favoring phagocytosis) in granulocytes. Overall, it is concluded that regardless of bacteria being alive or thermally inactivated, yogurt has common effects on the innate system, but the presence of live bacteria is necessary to achieve a potentiating effect on the specific immune response.


Sujet(s)
Yaourt , Humains , Méthode en double aveugle , Adulte , Mâle , Femelle , Études prospectives , Pasteurisation , Phagocytose , Cytokines/métabolisme , Jeune adulte , Immunoglobuline M/sang , Interféron gamma/métabolisme , Adulte d'âge moyen , Granulocytes/immunologie , Système immunitaire/effets des médicaments et des substances chimiques , Récepteurs du fragment Fc des IgG/métabolisme
2.
Invest Ophthalmol Vis Sci ; 65(6): 39, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38935032

RÉSUMÉ

Purpose: This study aimed to explore protective effects and potential mechanism of ectoine, a natural osmoprotectant, on ocular surface mucin production in dry eye disease. Methods: A dry eye model was established in C57BL/6 mice exposed to desiccating stress (DS) with untreated (UT) mice as controls. DS mice were topically treated with 2.0% ectoine or PBS vehicle. Corneal epithelial defects were assessed by Oregon Green Dextran (OGD) fluorescent staining. Conjunctival goblet cells, ocular mucins, and T help (Th) cytokines were evaluated by immunofluorescent staining or ELISA, and RT-qPCR. Results: Compared with UT mice, corneal epithelial defects were detected as strong punctate OGD fluorescent staining in DS mice with vehicle, whereas ectoine treatment largely reduced OGD staining to near-normal levels. Conjunctival goblet cell density and cell size decreased markedly in DS mice, but was significantly recovered by ectoine treatment. The protein production and mRNA expression of two gel-forming secreted MUC5AC and MUC2, and 4 transmembrane mucins, MUC1, MUC4, MUC16, and MUC15, largely decreased in DS mice, but was restored by ectoine. Furthermore, Th2 cytokine IL-13 was inhibited, whereas Th1 cytokine IFN-γ was stimulated at protein and mRNA levels in conjunctiva and draining cervical lymph nodes (CLNs) of DS mice, leading to decreased IL-13/IFN-γ ratio. Interestingly, 2.0% ectoine reversed their alternations and restored IL-13/IFN-γ balance. Conclusions: Our findings demonstrate that topical ectoine significantly reduces corneal damage, and enhances goblet cell density and mucin production through restoring imbalanced IL-13/IFN-γ signaling in murine dry eye model. This suggests therapeutic potential of natural osmoprotectant ectoine for dry eye disease.


Sujet(s)
Modèles animaux de maladie humaine , Syndromes de l'oeil sec , Cellules caliciformes , Interféron gamma , Interleukine-13 , Souris de lignée C57BL , Mucines , Animaux , Syndromes de l'oeil sec/métabolisme , Syndromes de l'oeil sec/traitement médicamenteux , Souris , Cellules caliciformes/métabolisme , Cellules caliciformes/effets des médicaments et des substances chimiques , Cellules caliciformes/anatomopathologie , Interféron gamma/métabolisme , Mucines/métabolisme , Mucines/biosynthèse , Mucines/génétique , Interleukine-13/métabolisme , Conjonctive/métabolisme , Conjonctive/effets des médicaments et des substances chimiques , Conjonctive/anatomopathologie , Test ELISA , Femelle , Épithélium antérieur de la cornée/métabolisme , Épithélium antérieur de la cornée/effets des médicaments et des substances chimiques , Réaction de polymérisation en chaine en temps réel , ARN messager/génétique , ARN messager/métabolisme , Acides aminés diaminés
3.
Sci Rep ; 14(1): 14586, 2024 06 25.
Article de Anglais | MEDLINE | ID: mdl-38918457

RÉSUMÉ

Natural killer (NK) cells play a key role in defense against Salmonella infections during the early phase of infection. Our previous work showed that the excretory/secretory products of Ascaris suum repressed NK activity in vitro. Here, we asked if NK cell functionality was influenced in domestic pigs during coinfection with Ascaris and Salmonella enterica serotype Typhimurium. Ascaris coinfection completely abolished the IL-12 and IL-18 driven elevation of IFN-γ production seen in CD16 + CD8α + perforin + NK cells of Salmonella single-infected pigs. Furthermore, Ascaris coinfection prohibited the Salmonella-driven rise in NK perforin levels and CD107a surface expression. In line with impaired effector functions, NK cells from Ascaris-single and coinfected pigs displayed elevated expression of the inhibitory KLRA1 and NKG2A receptors genes, contrasting with the higher expression of the activating NKp46 and NKp30 receptors in NK cells during Salmonella single infection. These differences were accompanied by the highly significant upregulation of T-bet protein expression in NK cells from Ascaris-single and Ascaris/Salmonella coinfected pigs. Together, our data strongly indicate a profound repression of NK functionality by an Ascaris infection which may hinder infected individuals from adequately responding to a concurrent bacterial infection.


Sujet(s)
Ascaridiose , Co-infection , Cellules tueuses naturelles , Maladies des porcs , Animaux , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Ascaridiose/immunologie , Ascaridiose/médecine vétérinaire , Ascaridiose/parasitologie , Co-infection/immunologie , Co-infection/microbiologie , Co-infection/parasitologie , Suidae , Maladies des porcs/parasitologie , Maladies des porcs/immunologie , Maladies des porcs/microbiologie , Salmonelloses animales/immunologie , Salmonella typhimurium/immunologie , Salmonella typhimurium/pathogénicité , Ascaris suum/immunologie , Interféron gamma/métabolisme , Perforine/métabolisme , Interleukine-12/métabolisme , Protéines à domaine boîte-T/métabolisme , Protéines à domaine boîte-T/génétique , Interleukine-18/métabolisme
4.
PLoS One ; 19(6): e0306117, 2024.
Article de Anglais | MEDLINE | ID: mdl-38923980

RÉSUMÉ

The development of a cancer vaccine has become an essential focus in the field of medical biotechnology and immunology. In our study, the NY-SAR-35 cancer/testis antigen was targeted to design a novel peptide vaccine using bioinformatics tools, and BALB/c mice were used to evaluate the vaccine's immunological function. This evaluation involved assessing peptide-specific IgG levels in the serum via ELISA and measuring the levels of IFN-γ, IL-4, and granzyme B in the supernatant of cultured splenocytes. The final vaccine construct consisted of two T lymphocyte epitopes linked by the AAY linker. This construct displayed high antigenicity, non-allergenicity, non-toxicity, stability, and ability to induce IFN-γ and IL-4. It showed stable dynamics with both human MHC-I and II molecules, as well as mouse MHC-II molecules, and revealed strong Van der Waals and electrostatic energies. Emulsifying our peptide vaccine in incomplete Freund's adjuvant resulted in a remarkable increase in the levels of IgG. The splenocytes of mice that received the combination of peptide and adjuvant displayed a noteworthy increase in IFN-γ, IL-4, and granzyme B secretion. Additionally, their lymphocytes exhibited higher proliferation rates compared to the control group. Our data demonstrated that our vaccine could stimulate a robust immune response, making it a promising candidate for cancer prevention. However, clinical trials are necessary to assess its efficacy in humans.


Sujet(s)
Antigènes néoplasiques , Tumeurs du sein , Vaccins anticancéreux , Biologie informatique , Souris de lignée BALB C , Vaccins sous-unitaires , Animaux , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/administration et posologie , Souris , Femelle , Antigènes néoplasiques/immunologie , Humains , Vaccins sous-unitaires/immunologie , Tumeurs du sein/immunologie , Déterminants antigéniques des lymphocytes T/immunologie , Interleukine-4/immunologie , Interféron gamma/immunologie , Interféron gamma/métabolisme , Immunoglobuline G/immunologie , Immunoglobuline G/sang , Granzymes , Modèles animaux de maladie humaine ,
5.
J Immunother Cancer ; 12(6)2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38925679

RÉSUMÉ

BACKGROUND: Despite continuous improvements in the new target and construction of chimeric antigen receptor (CAR)-T, relapse remains a significant challenge following CAR-T therapy. Tumor microenvironment (TME) strongly correlates with the efficacy of CAR-T therapy. V-domain Ig suppressor of T-cell activation (VISTA), which exerts a multifaceted and controversial role in regulating the TME, acts not only as a ligand on antigen-presenting cells but also functions as a receptor on T cells. However, the characteristics and underlying mechanisms governing endogenous T-cell activation by VISTA, which are pivotal for reshaping the TME, remain incompletely elucidated. METHODS: The immunocompetent B acute lymphoblastic leukemia (B-ALL), lymphoma, and melanoma murine models were employed to investigate the characteristics of endogenous T cells within the TME following CD19 and hCAIX CAR-T cell therapy, respectively. Furthermore, we examined the role of VISTA controlled by interferon (IFN)-γ signaling in regulating endogenous T-cell activation and functionality in B-ALL mice. RESULTS: We demonstrated that the administration of CD19 CAR-T or hCAIX CAR-T cell therapy elicited augmented immune responses of endogenous T cells within the TME of B-ALL, lymphoma, and melanoma mice, thereby substantiating the efficacy of CAR-T cell efficacy. However, in the TME lacking IFN-γ signaling, VISTA levels remained elevated, resulting in attenuated cytotoxicity of endogenous T cells and reduced B-ALL recipient survival. Mice treated with CD19 CAR-T cells exhibited increased proportions of endogenous memory T cells during prolonged remission, which possessed the tumor-responsive capabilities to protect against B-ALL re-challenge. Compared with wild-type (WT) CAR-T treated mice, the administration of IFN-γ-/- CAR-T to both WT and IFN-γ-/- recipients resulted in a reduction in the numbers of endogenous CD4+ and CD8+ effectors, while exhibiting increased populations of naïve-like CD4+ T and memory CD8+ T cells. VISTA expression consistently remained elevated in resting or memory CD4+ T cells, with distinct localization from programmed cell death protein-1 (PD-1) expressing T subsets. Blocking the VISTA signal enhanced dendritic cell-induced proliferation and cytokine production by syngeneic T cells. CONCLUSION: Our findings confirm that endogenous T-cell activation and functionality are regulated by VISTA, which is associated with the therapeutic efficiency of CAR-T and provides a promising therapeutic strategy for relapse cases in CAR-T therapy.


Sujet(s)
Interféron gamma , Animaux , Souris , Interféron gamma/métabolisme , Immunothérapie adoptive/méthodes , Antigènes CD19/métabolisme , Antigènes CD19/immunologie , Microenvironnement tumoral , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Humains , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Antigènes B7/métabolisme , Activation des lymphocytes , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/métabolisme , Protéines membranaires
6.
Nat Commun ; 15(1): 5403, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38926363

RÉSUMÉ

Idiopathic inflammatory myopathies (IIMs) are severe autoimmune diseases with poorly understood pathogenesis and unmet medical needs. Here, we examine the role of interferon γ (IFNγ) using NOD female mice deficient in the inducible T cell co-stimulator (Icos), which have previously been shown to develop spontaneous IFNγ-driven myositis mimicking human disease. Using muscle proteomic and spatial transcriptomic analyses we reveal profound myofiber metabolic dysregulation in these mice. In addition, we report muscle mitochondrial abnormalities and oxidative stress in diseased mice. Supporting a pathogenic role for oxidative stress, treatment with a reactive oxygen species (ROS) buffer compound alleviated myositis, preserved muscle mitochondrial ultrastructure and respiration, and reduced inflammation. Mitochondrial anomalies and oxidative stress were diminished following anti-IFNγ treatment. Further transcriptomic analysis in IIMs patients and human myoblast in vitro studies supported the link between IFNγ and mitochondrial dysfunction observed in mice. These results suggest that mitochondrial dysfunction, ROS and inflammation are interconnected in a self-maintenance loop, opening perspectives for mitochondria therapy and/or ROS targeting drugs in myositis.


Sujet(s)
Interféron gamma , Myosite , Stress oxydatif , Espèces réactives de l'oxygène , Animaux , Interféron gamma/métabolisme , Myosite/métabolisme , Myosite/anatomopathologie , Myosite/génétique , Humains , Femelle , Espèces réactives de l'oxygène/métabolisme , Souris , Souris de lignée NOD , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Modèles animaux de maladie humaine , Mitochondries du muscle/métabolisme , Mitochondries du muscle/anatomopathologie , Souris knockout , Myoblastes/métabolisme
7.
Int J Mol Sci ; 25(12)2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38928170

RÉSUMÉ

Reactive pustular eruptions (RPEs) can manifest in a variety of conditions, including pustular psoriasis (PP) and adult-onset immunodeficiency syndrome due to anti-interferon-γ autoantibody (AOID). These RPEs can be attributed to different causes, one of which is genetic factors. However, the genetic basis for pustular skin diseases remains poorly understood. In our study, we conducted whole-exome sequencing on a cohort of 17 AOID patients with pustular reactions (AOID-PR) and 24 PP patients. We found that 76% and 58% of the AOID-PR and PP patients, respectively, carried rare genetic variations within the filaggrin (FLG) gene family. A total of 12 out of 21 SNPs on FLG had previously received clinical classifications, with only p.Ser2706Ter classified as pathogenic. In contrast, none of the FLG3 SNPs identified in this study had prior clinical classifications. Overall, these variations had not been previously documented in cases of pustular disorders, and two of them were entirely novel discoveries. Immunohistochemical analysis of skin biopsies revealed that FLG variants like p.Ser860Trp, p.Gly3903Ter, p.Gly2440Glu, and p.Glu2133Asp caused reductions in FLG levels similar to the pathogenic FLG p.Ser2706Ter. These results highlight rare FLG variants as potential novel genetic risk factors contributing to pustule formation in both AOID and PP.


Sujet(s)
Asiatiques , Protéines filaggrine , Protéines de filaments intermédiaires , Polymorphisme de nucléotide simple , Humains , Protéines de filaments intermédiaires/génétique , Femelle , Mâle , Asiatiques/génétique , Adulte , Adulte d'âge moyen , , Prédisposition génétique à une maladie , Psoriasis/génétique , Psoriasis/anatomopathologie , Sujet âgé , Interféron gamma/génétique , Interféron gamma/métabolisme , Autoanticorps/immunologie , Peau/anatomopathologie , Peau/métabolisme
8.
Viruses ; 16(6)2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38932282

RÉSUMÉ

Porcine reproductive and respiratory syndrome virus (PRRSV) presents a significant threat to the global swine industry. The development of highly effective subunit nanovaccines is a promising strategy for preventing PRRSV variant infections. In this study, two different types of ferritin (Ft) nanovaccines targeting the major glycoprotein GP5, named GP5m-Ft and (Bp-IVp)3-Ft, were constructed and evaluated as vaccine candidates for PRRSV. Transmission electron microscopy (TEM) and dynamic light scattering (DLS) demonstrated that both purified GP5m-Ft and (Bp-IVp)3-Ft proteins could self-assemble into nanospheres. A comparison of the immunogenicity of GP5m-Ft and (Bp-IVp)3-Ft with an inactivated PRRSV vaccine in BALB/c mice revealed that mice immunized with GP5m-Ft exhibited the highest ELISA antibody levels, neutralizing antibody titers, the lymphocyte proliferation index, and IFN-γ levels. Furthermore, vaccination with the GP5m-Ft nanoparticle effectively protected piglets against a highly pathogenic PRRSV challenge. These findings suggest that GP5m-Ft is a promising vaccine candidate for controlling PRRS.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Ferritines , Souris de lignée BALB C , Nanoparticules , Syndrome dysgénésique et respiratoire porcin , Virus du syndrome respiratoire et reproducteur porcin , Protéines de l'enveloppe virale , Vaccins antiviraux , Animaux , Virus du syndrome respiratoire et reproducteur porcin/immunologie , Ferritines/immunologie , Suidae , Souris , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Nanoparticules/composition chimique , Syndrome dysgénésique et respiratoire porcin/prévention et contrôle , Syndrome dysgénésique et respiratoire porcin/immunologie , Vaccins antiviraux/immunologie , Vaccins antiviraux/administration et posologie , Protéines de l'enveloppe virale/immunologie , Protéines de l'enveloppe virale/génétique , Femelle , Interféron gamma/métabolisme ,
9.
Front Immunol ; 15: 1309846, 2024.
Article de Anglais | MEDLINE | ID: mdl-38919612

RÉSUMÉ

Acid ceramidase (Ac) is a lysosomal enzyme catalyzing the generation of sphingosine from ceramide, and Ac inhibitors are currently being investigated as potential cancer therapeutics. Yet, the role of the Ac in immune responses, particularly anti-viral immunity, is not fully understood. To investigate the impact of Ac expression on various leukocyte populations, we generated a tamoxifen-inducible global knockout mouse model for the Ac (iAc-KO). Following tamoxifen administration to healthy mice, we extracted primary and secondary lymphoid organs from iAc-KO and wild-type (wt) littermates and subsequently performed extensive flow cytometric marker analysis. In addition, we isolated CD4+ T cells from the spleen and lymph nodes for sphingolipid profiling and restimulated them in vitro with Dynabeads™ Mouse T-activator CD3/CD28. Intracellular cytokine expression (FACS staining) was analyzed and secreted cytokines detected in supernatants. To study cell-intrinsic effects, we established an in vitro model for iAc-KO in isolated CD4+ T and B cells. For CD4+ T cells of iAc-KO versus wt mice, we observed reduced Ac activity, an increased ceramide level, and enhanced secretion of IFNγ upon CD3/CD28 costimulation. Moreover, there was a marked reduction in B cell and plasma cell and blast numbers in iAc-KO compared to wt mice. To study cell-intrinsic effects and in line with the 3R principles, we established in vitro cell culture systems for iAc-KO in isolated B and CD4+ T cells. Our findings pinpoint to a key role of the Ac in mature B and antibody-secreting cells and in IFNγ secretion by CD4+ T cells.


Sujet(s)
Acid Ceramidase , Lymphocytes B , Lymphocytes T CD4+ , Interféron gamma , Souris knockout , Animaux , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Interféron gamma/métabolisme , Souris , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Acid Ceramidase/métabolisme , Acid Ceramidase/génétique , Souris de lignée C57BL , Numération des lymphocytes
10.
Nat Commun ; 15(1): 5318, 2024 Jun 22.
Article de Anglais | MEDLINE | ID: mdl-38909022

RÉSUMÉ

During primary varicella zoster virus (VZV) infection, infected lymphocytes drive primary viremia, causing systemic dissemination throughout the host, including the skin. This results in cytokine expression, including interferons (IFNs), which partly limit infection. VZV also spreads from skin keratinocytes to lymphocytes prior to secondary viremia. It is not clear how VZV achieves this while evading the cytokine response. Here, we show that VZV glycoprotein C (gC) binds IFN-γ and modifies its activity, increasing the expression of a subset of IFN-stimulated genes (ISGs), including intercellular adhesion molecule 1 (ICAM1), chemokines and immunomodulatory genes. The higher ICAM1 protein level at the plasma membrane of keratinocytes facilitates lymphocyte function-associated antigen 1-dependent T cell adhesion and expression of gC during infection increases VZV spread to peripheral blood mononuclear cells. This constitutes the discovery of a strategy to modulate IFN-γ activity, upregulating a subset of ISGs, promoting enhanced lymphocyte adhesion and virus spread.


Sujet(s)
Adhérence cellulaire , Herpèsvirus humain de type 3 , Molécule-1 d'adhérence intercellulaire , Interféron gamma , Kératinocytes , Lymphocytes T , Humains , Interféron gamma/métabolisme , Interféron gamma/immunologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Lymphocytes T/virologie , Molécule-1 d'adhérence intercellulaire/métabolisme , Molécule-1 d'adhérence intercellulaire/génétique , Kératinocytes/virologie , Kératinocytes/métabolisme , Kératinocytes/immunologie , Herpèsvirus humain de type 3/physiologie , Infection à virus varicelle-zona/immunologie , Infection à virus varicelle-zona/virologie , Agranulocytes/virologie , Agranulocytes/métabolisme , Agranulocytes/immunologie , Protéines de l'enveloppe virale/métabolisme , Antigène-1 associé à la fonction du lymphocyte/métabolisme
11.
Biomolecules ; 14(6)2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38927060

RÉSUMÉ

Plasmacytoid dendritic cells (pDCs) are vital players in antiviral immune responses because of their high levels of IFN-α secretion. However, this attribute has also implicated them as critical factors behind the immunopathogenesis of inflammatory diseases, and no currently available therapy can efficiently inhibit pDCs' aberrant activation. Mesenchymal stromal cells (MSCs) possess stromal immunomodulatory functionality, regulating immune cell activation through several mechanisms, including the adenosinergic (CD39/CD73/adenosine) pathway. The IFN-γ preconditioning of bone marrow MSCs improves their inhibitory properties for therapy applications; however, isolating human gingival tissue-derived MSCs (hGMSCs) is more accessible. These cells have shown better immunomodulatory effects, yet the outcome of IFN-γ preconditioning and its impact on the adenosinergic pathway has not been evaluated. This study first validated the immunoregulatory properties of primary-cultured hGMSCs, and the results showed that IFN-γ preconditioning strengthens CD39/CD73 coexpression, adenosine production, and the regulatory properties of hGMSC, which were confirmed by describing for the first time their ability to reduce pDC activation and their IFN-α secretion and to increase the frequency of CD73+ pDC. In addition, when CD73's enzymatic activity was neutralized in hGMSCs, adenosine production and the IFN-γ preconditioning effect were restrained. This evidence might be applied to design hGMSCs- and adenosine-based immunotherapeutic strategies for treating inflammatory disorders that are associated with pDC overactivation.


Sujet(s)
5'-Nucleotidase , Adénosine , Cellules dendritiques , Gencive , Interféron gamma , Cellules souches mésenchymateuses , Humains , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/immunologie , Cellules souches mésenchymateuses/cytologie , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Adénosine/métabolisme , Interféron gamma/métabolisme , Gencive/cytologie , 5'-Nucleotidase/métabolisme , Cellules cultivées , Apyrase/métabolisme , Protéines liées au GPI
12.
Immun Inflamm Dis ; 12(6): e1317, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38869352

RÉSUMÉ

BACKGROUND: Numerous studies have demonstrated that Absent in Melanoma 2 (AIM2) is upregulated in aortic plaques, especially in Vascular Smooth Muscle Cells in Coronary Artery Disease (CAD), and is related to inflammasome-induced inflammation. However, the underlying mechanism of this phenomenon and the role of AIM2 in atherosclerosis remained unclear. METHODS: This study enrolled 133 CAD patients and 123 controls. We isolated Peripheral Blood Leukocytes (PBLs) and the mRNA expression of AIM2 inflammasome and its downstream genes (ASC, Caspase-1, IL-1ß, and IL-18) were detected by real-time quantitative PCR (qPCR). We assessed correlations between AIM2 expressions and clinical characteristics by multiple linear regression and spearman's correlation. The THP-1 cells cultured in poly(dA:dT), A151, interferon-gamma (IFN-γ), AG490, or JC2-11. And then the mRNA and protein levels of AIM2, ASC, Caspase-1, IL-1ß, IL-18, GSDMD, and STAT1 were analyzed by qPCR and Western blot analysis, respectively. The migration and adhesive capacity of THP-1 cells was assessed using an inverted microscope and an inverted fluorescence microscope, respectively. RESULTS: In this study, we found that expressions of components of AIM2 inflammasome and its downstream genes (ASC, Caspase-1, IL-1ß, and IL-18), were all increased in PBLs of CAD patients, which indicated the inflammasome activation. AIM2 inflammasome activation further induced pyroptosis, and stimulated migration and adhesion in monocyte cell lines, which was regulated by IFN-γ probably through JAK2/STAT1 pathway. In addition, AIM2 expressions were positively correlated with systemic inflammatory indicators as an independent risk factor for CAD. CONCLUSIONS: In conclusion, increased AIM2 expression, induced by the IFN-γ/JAK2/STAT1 signal, orientates monocytes to inflammatory status or even pyroptosis through AIM2 inflammasome activation, which is involved in the development of CAD.


Sujet(s)
Maladie des artères coronaires , Protéines de liaison à l'ADN , Inflammasomes , Interféron gamma , Kinase Janus-2 , Monocytes , Pyroptose , Facteur de transcription STAT-1 , Transduction du signal , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Maladie des artères coronaires/immunologie , Maladie des artères coronaires/métabolisme , Maladie des artères coronaires/anatomopathologie , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Inflammasomes/métabolisme , Interféron gamma/métabolisme , Kinase Janus-2/métabolisme , Kinase Janus-2/génétique , Monocytes/métabolisme , Monocytes/immunologie , Facteur de transcription STAT-1/métabolisme , Cellules THP-1
13.
J Sports Sci ; 42(8): 751-762, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38864405

RÉSUMÉ

Few studies have reported the cardiovascular health effects of different high-intensity interval training (HIIT) protocols among sedentary young women. We investigated the impact of a traditional HIIT programme and a high-intensity circuit training (HICT) programme on lipid profiles and inflammatory cytokine levels in sedentary young women. Forty-two women were randomly assigned to HICT (body weight-based training), HIIT (cycling-based training), or control groups (n = 14 each). HICT and HIIT participants completed an 8-week training programme of three sessions per week. Total cholesterol (TC), triglyceride, high- and low-density lipoprotein, leptin, resistin, tumour necrosis factor-alpha (TNF-α), interleukin-8, and interferon-gamma levels were measured before and after the intervention. Post-intervention, TC and leptin were decreased in the HICT group. The HICT group also demonstrated increased lean mass, upper and lower limb strength, and balance, while the HIIT group displayed improved lower limb strength. Additionally, the control group showed significant increases in triglyceride levels, weight, body mass index, and fat mass. In conclusion, although both HICT and HIIT interventions showed improvements in cardiovascular health and physical fitness, participants in the HICT group experienced more health benefits.


Sujet(s)
Marqueurs biologiques , Entrainement fractionné de haute intensité , Leptine , Mode de vie sédentaire , Humains , Entrainement fractionné de haute intensité/méthodes , Femelle , Marqueurs biologiques/sang , Leptine/sang , Jeune adulte , Triglycéride/sang , Indice de masse corporelle , Facteur de nécrose tumorale alpha/sang , Lipides/sang , Force musculaire/physiologie , Composition corporelle , Résistine/sang , Cytokines/sang , Cholestérol/sang , Adulte , Interféron gamma/sang , Interleukine-8/sang
14.
Front Immunol ; 15: 1345046, 2024.
Article de Anglais | MEDLINE | ID: mdl-38827732

RÉSUMÉ

Introduction: Recently, more and more research illustrated the importance of inducing CD4+ T helper type (Th)-1 dominant immunity for the success of tumor immunotherapy. Our prior studies revealed the crucial role of CD4+ Th1 cells in orchestrating systemic and durable antitumor immunity, which contributes to the satisfactory outcomes of the novel cryo-thermal therapy in the B16F10 tumor model. However, the mechanism for maintaining the cryo-thermal therapy-mediated durable CD4+ Th1-dominant response remains uncovered. Additionally, cryo-thermal-induced early-stage CD4+ Th1-dominant T cell response showed a correlation with the favorable prognosis in patients with colorectal cancer liver metastasis (CRCLM). We hypothesized that CD4+ Th1-dominant differentiation induced during the early stage post cryo-thermal therapy would affect the balance of CD4+ subsets at the late phase. Methods: To understand the role of interferon (IFN)-γ, the major effector of Th1 subsets, in maintaining long-term CD4+ Th1-prone polarization, B16F10 melanoma model was established in this study and a monoclonal antibody was used at the early stage post cryo-thermal therapy for interferon (IFN)-γ signaling blockade, and the influence on the phenotypic and functional change of immune cells was evaluated. Results: IFNγ at the early stage after cryo-thermal therapy maintained long-lasting CD4+ Th1-prone immunity by directly controlling Th17, Tfh, and Tregs polarization, leading to the hyperactivation of Myeloid-derived suppressor cells (MDSCs) represented by abundant interleukin (IL)-1ß generation, and thereby further amplifying Th1 response. Discussion: Our finding emphasized the key role of early-phase IFNγ abundance post cryo-thermal therapy, which could be a biomarker for better prognosis after cryo-thermal therapy.


Sujet(s)
Différenciation cellulaire , Interféron gamma , Mélanome expérimental , Souris de lignée C57BL , Lymphocytes auxiliaires Th1 , Animaux , Lymphocytes auxiliaires Th1/immunologie , Souris , Interféron gamma/métabolisme , Différenciation cellulaire/immunologie , Mélanome expérimental/immunologie , Mélanome expérimental/thérapie , Cryothérapie/méthodes , Lignée cellulaire tumorale , Femelle
15.
Commun Biol ; 7(1): 699, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38849594

RÉSUMÉ

Caspase-4 (CASP4) is a member of the inflammatory caspase subfamily and promotes inflammation. Here, we report that CASP4 in lung adenocarcinoma cells contributes to both tumor progression via angiogenesis and tumor hyperkinesis and tumor cell killing in response to high interferon (IFN)-γ levels. We observe that elevated CASP4 expression in the primary tumor is associated with cancer progression in patients with lung adenocarcinoma. Further, CASP4 knockout attenuates tumor angiogenesis and metastasis in subcutaneous tumor mouse models. CASP4 enhances the expression of genes associated with angiogenesis and cell migration in lung adenocarcinoma cell lines through nuclear factor kappa-light chain-enhancer of activated B cell signaling without stimulation by lipopolysaccharide or tumor necrosis factor. CASP4 is induced by endoplasmic reticulum stress or IFN-γ via signal transducer and activator of transcription 1. Most notably, lung adenocarcinoma cells with high CASP4 expression are more prone to IFN-γ-induced pyroptosis than those with low CASP4 expression. Our findings indicate that the CASP4 level in primary lung adenocarcinoma can predict metastasis and responsiveness to high-dose IFN-γ therapy due to cancer cell pyroptosis.


Sujet(s)
Adénocarcinome pulmonaire , Caspases initiatrices , Interféron gamma , Tumeurs du poumon , Pyroptose , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Animaux , Interféron gamma/métabolisme , Interféron gamma/pharmacologie , Interféron gamma/génétique , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Souris , Caspases initiatrices/métabolisme , Caspases initiatrices/génétique , Lignée cellulaire tumorale , Métastase tumorale , Régulation de l'expression des gènes tumoraux
16.
Iran J Allergy Asthma Immunol ; 23(2): 197-220, 2024 Apr 07.
Article de Anglais | MEDLINE | ID: mdl-38822514

RÉSUMÉ

Systemic sclerosis (SSc) is an autoimmune systemic disease that is characterized by immune dysregulation, inflammation, vasculopathy, and fibrosis. Tissue fibrosis plays an important role in SSc and can affect several organs such as the dermis, lungs, and heart. Dysregulation of interferon (IFN) signaling contributes to the SSc pathogenesis and interferon regulatory factor 1 (IRF1) has been indicated as the main regulator of type I IFN. This study aimed to clarify the effect of IFN-gamma (-γ) and dexamethasone (DEX) on the IRF1, extracellular signal-regulated kinase 1/2 (ERK1/2), and the expression of alpha-smooth muscle actin (α-SMA) in myofibroblasts and genes involved in the inflammation and fibrosis processes in early diffuse cutaneous systemic sclerosis (dcSSc). A total of 10 early dcSSc patients (diffuse cutaneous form) and 10 unaffected control dermis biopsies were obtained to determine IFNγ and DEX effects on inflammation and fibrosis. Fibroblasts were treated with IFNγ and DEX at optimum time and dose. The expression level of genes and proteins involved in the fibrosis and inflammation processes have been quantified by quantitative real-time PCR (RT-qPCR) and western blot, respectively. IFNγ could up-regulate some of the inflammation-related genes (Interleukin-6; IL6) and down-regulate some of the fibrosis-related genes (COL1A1) in cultured fibroblasts of patients with early dcSSc compared to the untreated group. Besides, it has been revealed that IFNγ can induce fibroblast differentiation to the myofibroblast that expresses α-SMA. Concerning the inhibitory effect of IFNγ on some fibrotic genes and its positive effect on the inflammatory genes and myofibroblast differentiation, it seems that IFNγ may play a dual role in SSc.


Sujet(s)
Actines , Fibroblastes , Interféron gamma , Interleukine-6 , Sclérodermie systémique , Adulte , Femelle , Humains , Mâle , Adulte d'âge moyen , Actines/métabolisme , Actines/génétique , Cellules cultivées , Dexaméthasone/pharmacologie , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Fibroblastes/effets des médicaments et des substances chimiques , Fibrose , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Facteur-1 de régulation d'interféron/métabolisme , Facteur-1 de régulation d'interféron/génétique , Interféron gamma/pharmacologie , Interleukine-6/métabolisme , Interleukine-6/génétique , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Sclérodermie systémique/métabolisme , Sclérodermie systémique/anatomopathologie , Sclérodermie systémique/immunologie
17.
Vet Res ; 55(1): 69, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38822400

RÉSUMÉ

Current diagnostic methods for Johne's disease in cattle allow reliable detection of infections with Mycobacterium avium ssp. paratuberculosis (MAP) not before animals are 2 years of age. Applying a flow cytometry-based approach (FCA) to quantify a MAP-specific interferon-gamma (IFN-γ) response in T cell subsets, the present study sought to monitor the kinetics of the cell-mediated immune response in experimentally infected calves. Six MAP-negative calves and six calves, orally inoculated with MAP at 10 days of age, were sampled every 4 weeks for 52 weeks post-inoculation (wpi). Peripheral blood mononuclear cells (PBMC) were stimulated with either purified protein derivatives (PPD) or whole cell sonicates derived from MAP (WCSj), M. avium ssp. avium or M. phlei for 6 days followed by labeling of intracellular IFN-γ in CD4+ and CD8+ T cells. No antigen-specific IFN-γ production was detectable in CD8+ cells throughout and the responses of CD4+ cells of MAP-infected and control calves were similar up to 12 wpi. However, the mean fluorescence intensity (MFI) for the detection of IFN-γ in CD4+ cells after WCSj antigen stimulation allowed for a differentiation of animal groups from 16 wpi onwards. This approach had a superior sensitivity (87.8%) and specificity (86.8%) to detect infected animals from 16 wpi onwards, i.e., in an early infection stage, as compared to the IFN-γ release assay (IGRA). Quantification of specific IFN-γ production at the level of individual CD4+ cells may serve, therefore, as a valuable tool to identify MAP-infected juvenile cattle.


Sujet(s)
Lymphocytes T CD4+ , Maladies des bovins , Cytométrie en flux , Interféron gamma , Mycobacterium avium ssp. paratuberculosis , Paratuberculose , Animaux , Bovins , Paratuberculose/immunologie , Paratuberculose/diagnostic , Paratuberculose/microbiologie , Mycobacterium avium ssp. paratuberculosis/immunologie , Mycobacterium avium ssp. paratuberculosis/physiologie , Interféron gamma/métabolisme , Cytométrie en flux/médecine vétérinaire , Cytométrie en flux/méthodes , Maladies des bovins/immunologie , Maladies des bovins/diagnostic , Maladies des bovins/microbiologie , Lymphocytes T CD4+/immunologie , Marqueurs biologiques
18.
PLoS One ; 19(6): e0299631, 2024.
Article de Anglais | MEDLINE | ID: mdl-38870172

RÉSUMÉ

Inflammation-derived oxidative stress is postulated to contribute to neuronal damage leading to poor clinical outcomes in Acute Ischemic Stroke (AIS). We aimed to investigate the association between serum levels of selected cytokines (IL-1ß, IFN-γ, IL-4), and vitamin D in ischemic stroke progression, and their accuracy in predicting AIS prognosis, among Sri Lankans. We compared 60 AIS patients admitted in 4 phases post-stroke onset (<6 h; 6-24 h; 24-48 h; 48-96 h; n = 15/phase), with 15 age- and sex-matched controls. The 30-day functional outcome (FO) was assessed using the modified Rankin Scale (mRS). Serum cytokine and vitamin D levels were quantified using sandwich ELISAs, and competitive ELISA, respectively. The CombiROC web tool established optimal prognostic biomarker combinations. Serum IL-1ß and IFN-γ were elevated in all four phases following stroke onset while IL-4 was elevated exclusively in the recovery phase (48-96 h) (p<0.05). Th1 bias polarization of the Th1:Th2 cytokine (IFN-γ:IL-4) ratio occurred with AIS progression while a Th2 bias occurred during AIS recovery (p<0.05). Lower serum IL-1ß and higher IL-4 levels were associated with a good FO (p<0.05), while lower Vitamin D levels were related to a poor FO (p = 0.001). The triple-biomarker panel, IL-4- IFN-γ -Vit D, accurately predicted AIS prognosis (sensitivity = 100%, specificity = 91.9%, area under the curve = 0.98). Serum immunologic mediators IFN-γ, IL-4, and vitamin D may be useful biomarkers of AIS prognosis and may serve as therapeutic targets in improving stroke outcomes. Vitamin D supplementation may improve the prognosis of AIS patients. Furthermore, binary logistic model fitted for FO indicated Th1:Th2 cytokine ratio (IFN-γ:IL-4), vitamin D status, history of stroke, and ischemic heart disease as significant predictors of AIS prognosis.


Sujet(s)
Marqueurs biologiques , Cytokines , Accident vasculaire cérébral ischémique , Vitamine D , Humains , Femelle , Mâle , Vitamine D/sang , Accident vasculaire cérébral ischémique/sang , Accident vasculaire cérébral ischémique/diagnostic , Pronostic , Marqueurs biologiques/sang , Adulte d'âge moyen , Sujet âgé , Cytokines/sang , Interleukine-4/sang , Interféron gamma/sang , Études cas-témoins
19.
Front Immunol ; 15: 1394114, 2024.
Article de Anglais | MEDLINE | ID: mdl-38873610

RÉSUMÉ

Introduction: Several effective vaccines for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been developed and implemented in the population. However, the current production capacity falls short of meeting global demand. Therefore, it is crucial to further develop novel vaccine platforms that can bridge the distribution gap. AVX/COVID-12 is a vector-based vaccine that utilizes the Newcastle Disease virus (NDV) to present the SARS-CoV-2 spike protein to the immune system. Methods: This study aims to analyze the antigenicity of the vaccine candidate by examining antibody binding and T-cell activation in individuals infected with SARS-CoV-2 or variants of concern (VOCs), as well as in healthy volunteers who received coronavirus disease 2019 (COVID-19) vaccinations. Results: Our findings indicate that the vaccine effectively binds antibodies and activates T-cells in individuals who received 2 or 3 doses of BNT162b2 or AZ/ChAdOx-1-S vaccines. Furthermore, the stimulation of T-cells from patients and vaccine recipients with AVX/COVID-12 resulted in their proliferation and secretion of interferon-gamma (IFN-γ) in both CD4+ and CD8+ T-cells. Discussion: The AVX/COVID-12 vectored vaccine candidate demonstrates the ability to stimulate robust cellular responses and is recognized by antibodies primed by the spike protein present in SARS-CoV-2 viruses that infected patients, as well as in the mRNA BNT162b2 and AZ/ChAdOx-1-S vaccines. These results support the inclusion of the AVX/COVID-12 vaccine as a booster in vaccination programs aimed at addressing COVID-19 caused by SARS-CoV-2 and its VOCs.


Sujet(s)
Anticorps antiviraux , Vaccins contre la COVID-19 , COVID-19 , Activation des lymphocytes , Virus de la maladie de Newcastle , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Humains , COVID-19/immunologie , COVID-19/prévention et contrôle , SARS-CoV-2/immunologie , Anticorps antiviraux/immunologie , Virus de la maladie de Newcastle/immunologie , Vaccins contre la COVID-19/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Activation des lymphocytes/immunologie , Adulte , Femelle , Mâle , Adulte d'âge moyen , Lymphocytes T/immunologie , Vaccin BNT162/immunologie , Vaccination , Vecteurs génétiques/génétique , Vecteurs génétiques/immunologie , Interféron gamma/immunologie , Interféron gamma/métabolisme
20.
Clin Exp Hypertens ; 46(1): 2366270, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-38864268

RÉSUMÉ

OBJECTIVE: To elucidate the underlying mechanism by which the proliferation and migration abilities of human umbilical cord mesenchymal stem cells (hUC-MSCs) determine their therapeutic efficacy in rheumatoid arthritis treatment. METHODS: The DBA/1J mice were utilized to establish a collagen-induced RA (CIA) mouse model and to validate the therapeutic efficacy of hUC-MSCs transfected with CD151 siRNA. RNA-seq, QT-PCR and western blotting were utilized to evaluate the mRNA and protein levels of the PI3K/AKT pathway, respectively. RESULTS: IFN-γ significantly enhanced the proliferation and migration abilities of hUC-MSCs, up-regulating the expression of CD151, a gene related to cell proliferation and migration. Effective inhibition of this effect was achieved through CD151 siRNA treatment. However, IFN-γ did not affect hUC-MSCs differentiation or changes in cell surface markers. Additionally, transplantation of CD151-interfered hUC-MSCs (siRNA-CD151-hUC-MSCs) resulted in decreased colonization in the toes of CIA mice and worse therapeutic effects compared to empty vector treatment (siRNA-NC-hUC-MSCs). CONCLUSION: IFN-γ facilitates the proliferation and migration of hUC-MSCs through the CD151/PI3K/AKT pathway. The therapeutic efficacy of siRNA-CD151-hUC-MSCs was found to be inferior to that of siRNA-NC-hUC-MSCs.


Sujet(s)
Polyarthrite rhumatoïde , Mouvement cellulaire , Prolifération cellulaire , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Souris de lignée DBA , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Animaux , Polyarthrite rhumatoïde/thérapie , Polyarthrite rhumatoïde/métabolisme , Souris , Cellules souches mésenchymateuses/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Transplantation de cellules souches mésenchymateuses/méthodes , Phosphatidylinositol 3-kinases/métabolisme , Humains , Interféron gamma/métabolisme , Cordon ombilical/cytologie , Arthrite expérimentale/thérapie , Arthrite expérimentale/métabolisme , Mâle
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...