Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 22.289
Filtrer
1.
J Immunol Res ; 2024: 5537948, 2024.
Article de Anglais | MEDLINE | ID: mdl-39056014

RÉSUMÉ

CD8+ T cells are essential for adaptive immunity against infection and tumors. Their ability to proliferate after stimulation is crucial to their functionality. Dendritic cells (DCs) are professional antigen-presenting cells that induce their proliferation. Here, we show that thapsigargin-induced LAD2 mast cell (MC) line-released products can impair the ability of monocyte-derived DCs to induce CD8+ T-cell proliferation and the generation of Th1 cytokine-producing T cells. We found that culture medium conditioned with LAD2 MCs previously stimulated with thapsigargin (thapsLAD2) induces maturation of DCs as determined by the maturation markers CD80, CD83, CD86, and HLA-DR. However, thapsLAD2-matured DCs produced no detectable TNFα or IL-12 during the maturation. In addition, although their surface expression of PD-L1 was comparable with the immature or TLR7/8-agonist (R848)-matured DCs, their TIM-3 expression was significantly higher than in immature DCs and even much higher than in R848-matured DCs. In addition, contrary to R848-matured DCs, the thapsLAD2-matured DCs only tended to induce enhanced proliferation of CD4+ T cells than immature DCs. For CD8+ T cells, this tendency was not even detected because thapsLAD2-matured and immature DCs comparably induced their proliferation, which contrasted with the significantly enhanced proliferation induced by R848-matured DCs. Furthermore, these differences were comparably recapitulated in the ability of the tested DCs to induce IFNγ- and IFNγ/TNFα-producing T cells. These findings show a novel mechanism of MC-mediated regulation of adaptive immune responses.


Sujet(s)
Lymphocytes T CD8+ , Différenciation cellulaire , Prolifération cellulaire , Cellules dendritiques , Activation des lymphocytes , Mastocytes , Thapsigargine , Humains , Cellules dendritiques/immunologie , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/métabolisme , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Thapsigargine/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Mastocytes/immunologie , Mastocytes/effets des médicaments et des substances chimiques , Mastocytes/métabolisme , Activation des lymphocytes/effets des médicaments et des substances chimiques , Activation des lymphocytes/immunologie , Monocytes/immunologie , Monocytes/effets des médicaments et des substances chimiques , Monocytes/métabolisme , Récepteur cellulaire-2 du virus de l'hépatite A/métabolisme , Cytokines/métabolisme , Imidazoles/pharmacologie , Lignée cellulaire
2.
BMC Immunol ; 25(1): 48, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39054418

RÉSUMÉ

BACKGROUND: TLR7 is a key player in the antiviral immunity. TLR7 signaling activates antigen-presenting cells including DCs and macrophages. This activation results in the adaptive immunity including T cells and B cells. Therefore, TLR7 is an important molecule of the immune system. Based on these observations, TLR7 agonists considered to become a therapy weaponize the immune system against cancer. Radiation therapy (RT) is one of the standard cancer therapies and is reported to modulate the tumor immune response. In this study, we aimed to investigate the anti-tumor activity in combination of TLR7 agonist, DSP-0509, with RT and underlying mechanism. RESULT: We showed that anti-tumor activity is enhanced by combining RT with the TLR7 agonist DSP-0509 in the CT26, LM8, and 4T1 inoculated mice models. We found that once- weekly (q1w) dosing of DSP-0509 rather than biweekly (q2w) dosing is needed to achieve superior anti-tumor activities in CT26 model. Spleen cells from the mice in RT/DSP-0509 combination treatment group showed increased tumor lytic activity, inversely correlated with tumor volume, as measured by the chromium-release cytotoxicity assay. We also found the level of cytotoxic T lymphocytes (CTLs) increased in the spleens of completely cured mice. When the mice completely cured by combination therapy were re-challenged with CT26 cells, all mice rejected CT26 cells but accepted Renca cells. This rejection was not observed with CD8 depletion. Furthermore, levels of splenic effector memory CD8 T cells were increased in the combination therapy group. To explore the factors responsible for complete cure by combination therapy, we analyzed peripheral blood leukocytes (PBLs) mRNA from completely cured mice. We found that Havcr2low, Cd274low, Cd80high, and Il6low were a predictive signature for the complete response to combination therapy. An analysis of tumor-derived mRNA showed that combination of RT and DSP-0509 strongly increased the expression of anti-tumor effector molecules including Gzmb and Il12. CONCLUSION: These data suggest that TLR7 agonist, DSP-0509, can be a promising concomitant when used in combination with RT by upregulating CTLs activity and gene expression of effector molecules. This combination can be an expecting new radio-immunotherapeutic strategy in clinical trials.


Sujet(s)
Récepteur de type Toll-7 , Animaux , Récepteur de type Toll-7/agonistes , Souris , Lignée cellulaire tumorale , Femelle , Activation des lymphocytes/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Glycoprotéines membranaires/agonistes , Association thérapeutique , Humains , Souris de lignée C57BL , Tumeurs/thérapie , Tumeurs/immunologie , Tumeurs/traitement médicamenteux , Modèles animaux de maladie humaine , Lymphocytes T/immunologie , Lymphocytes T/effets des médicaments et des substances chimiques
3.
Int J Mol Sci ; 25(13)2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-39000566

RÉSUMÉ

Staphylococcal toxic shock syndrome (STSS) is a rare, yet potentially fatal disease caused by Staphylococcus aureus (S. aureus) enterotoxins, known as superantigens, which trigger an intense immune response. Our previous study demonstrated the protective effect of tofacitinib against murine toxin-induced shock and a beneficial effect against S. aureus sepsis. In the current study, we examined the effects of tofacitinib on T-cell response in peripheral blood using a mouse model of enterotoxin-induced shock. Our data revealed that tofacitinib suppresses the activation of both CD4+ and CD8+ T cells in peripheral blood. Furthermore, both gene and protein levels of Th1 cytokines were downregulated by tofacitinib treatment in mice with enterotoxin-induced shock. Importantly, we demonstrated that CD4+ cells, but not CD8+ cells, are pathogenic in mice with enterotoxin-induced shock. In conclusion, our findings suggest that tofacitinib treatment suppresses CD4+ T-cell activation and Th1 response, thereby aiding in protection against staphylococcal toxic shock in mice. This insight may guide the future development of novel therapies for STSS.


Sujet(s)
Lymphocytes T CD4+ , Activation des lymphocytes , Pipéridines , Pyrimidines , Choc septique , Infections à staphylocoques , Lymphocytes auxiliaires Th1 , Animaux , Pipéridines/pharmacologie , Pipéridines/usage thérapeutique , Lymphocytes auxiliaires Th1/immunologie , Lymphocytes auxiliaires Th1/effets des médicaments et des substances chimiques , Lymphocytes auxiliaires Th1/métabolisme , Pyrimidines/pharmacologie , Pyrimidines/usage thérapeutique , Choc septique/traitement médicamenteux , Choc septique/immunologie , Choc septique/induit chimiquement , Souris , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Lymphocytes T CD4+/métabolisme , Activation des lymphocytes/effets des médicaments et des substances chimiques , Infections à staphylocoques/traitement médicamenteux , Infections à staphylocoques/immunologie , Infections à staphylocoques/microbiologie , Entérotoxines , Staphylococcus aureus/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Souris de lignée C57BL , Femelle , Modèles animaux de maladie humaine , Superantigènes/immunologie
4.
Int J Mol Sci ; 25(13)2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-39000607

RÉSUMÉ

Natural killer (NK) cells play a crucial role in innate immunity, particularly in combating infections and tumors. However, in hematological cancers, NK cells often exhibit impaired functions. Therefore, it is very important to activate its endosomal Toll-like receptors (TLRs) as a potential strategy to restore its antitumor activity. We stimulated NK cells from the peripheral blood mononuclear cells from children with acute lymphoblastic leukemia and NK cells isolated, and the NK cells were stimulated with specific TLR ligands (Poly I:C, Imiquimod, R848, and ODN2006) and we evaluated changes in IFN-γ, CD107a, NKG2D, NKp44 expression, Granzyme B secretion, cytokine/chemokine release, and cytotoxic activity. Results revealed that Poly I:C and Imiquimod enhanced the activation of both immunoregulatory and cytotoxic NK cells, increasing IFN-γ, CD107a, NKG2D, and NKp44 expression. R848 activated immunoregulatory NK cells, while ODN2006 boosted CD107a, NKp44, NKG2D, and IFN-γ secretion in cytotoxic NK cells. R848 also increased the secretion of seven cytokines/chemokines. Importantly, R848 and ODN 2006 significantly improved cytotoxicity against leukemic cells. Overall, TLR stimulation enhances NK cell activation, suggesting TLR8 (R848) and TLR9 (ODN 2006) ligands as promising candidates for antitumor immunotherapy.


Sujet(s)
Imiquimod , Cellules tueuses naturelles , Activation des lymphocytes , Poly I-C , Leucémie-lymphome lymphoblastique à précurseurs B et T , Récepteurs de type Toll , Humains , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/effets des médicaments et des substances chimiques , Cellules tueuses naturelles/métabolisme , Leucémie-lymphome lymphoblastique à précurseurs B et T/immunologie , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B et T/anatomopathologie , Poly I-C/pharmacologie , Activation des lymphocytes/effets des médicaments et des substances chimiques , Activation des lymphocytes/immunologie , Imiquimod/pharmacologie , Récepteurs de type Toll/métabolisme , Récepteurs de type Toll/agonistes , Enfant , Oligodésoxyribonucléotides/pharmacologie , Cytokines/métabolisme , Femelle , Interféron gamma/métabolisme , Mâle , Imidazoles/pharmacologie , Cytotoxicité immunologique/effets des médicaments et des substances chimiques , Enfant d'âge préscolaire ,
5.
Arthritis Res Ther ; 26(1): 134, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-39020445

RÉSUMÉ

OBJECTIVE: To investigate the distribution and activation of B-cell subpopulations in rheumatoid arthritis (RA) patients treated with Janus kinase inhibitors (JAKis) and to analyze their correlation with disease remission. METHODS: Peripheral blood samples were collected from 23 adult healthy controls and 58 RA patients, 31 of whom were treated with JAKis and assessed during a 24-month follow-up. The number of peripheral B-cell subpopulations (including naive B cells, nonswitched memory B (NSMB) cells, switched memory B cells, and double-negative B cells), their activation, and phosphorylation of SYK and AKT upon B-cell receptor (BCR) stimulation in each population were analyzed by flow cytometry. RESULTS: Compared with that in healthy controls, the frequency of NSMB cells was significantly lower in new-onset untreated RA patients. However, expression of CD40, CD80, CD95, CD21low and pAKT significantly increased in these NSMB cells. Additionally, the number of NSMB cells correlated negatively with DAS28-ESR and IgG and IgA levels in these patients; expression of CD80, CD95 and CD21low on NSMB cells correlated positively with DAS28-ESR and IgG and IgA levels. After treatment with JAKis, the serum IgG concentration significantly decreased in RA patients in remission, but CD40, CD95 and pAKT levels in NSMB cells significantly decreased. CONCLUSION: RA patients present different B-cell subpopulations, in which the frequency of NSMB cells is negatively associated with disease activity. However, treatment with JAKis can inhibit activation of NSMB cells, restore the balance of kinase phosphorylation, and facilitate disease remission in RA patients.


Sujet(s)
Polyarthrite rhumatoïde , Inhibiteurs des Janus kinases , Humains , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/sang , Mâle , Adulte d'âge moyen , Femelle , Inhibiteurs des Janus kinases/usage thérapeutique , Inhibiteurs des Janus kinases/pharmacologie , Adulte , Cellules B mémoire/immunologie , Cellules B mémoire/effets des médicaments et des substances chimiques , Induction de rémission , Sujet âgé , Activation des lymphocytes/effets des médicaments et des substances chimiques , Activation des lymphocytes/immunologie , Antirhumatismaux/usage thérapeutique , Cytométrie en flux , Lymphocytes B/immunologie , Lymphocytes B/effets des médicaments et des substances chimiques , Lymphocytes B/métabolisme
6.
Oncoimmunology ; 13(1): 2373526, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948931

RÉSUMÉ

Prostate cancer (PCa) is characterized as a "cold tumor" with limited immune responses, rendering the tumor resistant to immune checkpoint inhibitors (ICI). Therapeutic messenger RNA (mRNA) vaccines have emerged as a promising strategy to overcome this challenge by enhancing immune reactivity and significantly boosting anti-tumor efficacy. In our study, we synthesized Tetra, an mRNA vaccine mixed with multiple tumor-associated antigens, and ImmunER, an immune-enhancing adjuvant, aiming to induce potent anti-tumor immunity. ImmunER exhibited the capacity to promote dendritic cells (DCs) maturation, enhance DCs migration, and improve antigen presentation at both cellular and animal levels. Moreover, Tetra, in combination with ImmunER, induced a transformation of bone marrow-derived dendritic cells (BMDCs) to cDC1-CCL22 and up-regulated the JAK-STAT1 pathway, promoting the release of IL-12, TNF-α, and other cytokines. This cascade led to enhanced proliferation and activation of T cells, resulting in effective killing of tumor cells. In vivo experiments further revealed that Tetra + ImmunER increased CD8+T cell infiltration and activation in RM-1-PSMA tumor tissues. In summary, our findings underscore the promising potential of the integrated Tetra and ImmunER mRNA-LNP therapy for robust anti-tumor immunity in PCa.


Sujet(s)
Adjuvants immunologiques , Antigènes néoplasiques , Vaccins anticancéreux , Cellules dendritiques , Tumeurs de la prostate , ARN messager , Animaux , Mâle , Tumeurs de la prostate/immunologie , Tumeurs de la prostate/thérapie , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Tumeurs de la prostate/traitement médicamenteux , Antigènes néoplasiques/immunologie , Souris , Cellules dendritiques/immunologie , Adjuvants immunologiques/pharmacologie , Adjuvants immunologiques/administration et posologie , ARN messager/génétique , ARN messager/métabolisme , ARN messager/administration et posologie , Vaccins anticancéreux/administration et posologie , Vaccins anticancéreux/immunologie , Humains , Souris de lignée C57BL , Lignée cellulaire tumorale , Vaccins à ARNm , Lymphocytes T CD8+/immunologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Immunothérapie/méthodes , Activation des lymphocytes/effets des médicaments et des substances chimiques
7.
Arch Dermatol Res ; 316(7): 348, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38849562

RÉSUMÉ

This study investigates the mechanism through which paeoniflorin inhibits TSLP expression to regulate dendritic cell activation in corticosteroid-dependent dermatitis treatment. Utilizing databases like TCMSP, we identified paeoniflorin's components, targets, and constructed networks. Molecular docking and gene enrichment analysis helped pinpoint key targets and pathways affected by paeoniflorin. In vitro and in vivo models were used to study CD80, CD86, cytokines, T-cell activation, skin lesions, histopathological changes, TSLP, CD80, and CD86 expression. Our study revealed paeoniflorin's active constituent targeting IL-6 in corticosteroid-dependent dermatitis. In vitro experiments demonstrated reduced TSLP expression, CD80, CD86, and cytokine secretion post-paeoniflorin treatment. In vivo, paeoniflorin significantly decreased skin lesion severity, cytokine levels, TSLP, CD80, and CD86 expression. The study highlights paeoniflorin's efficacy in inhibiting TSLP expression and suppressing dendritic cell activation in corticosteroid-dependent dermatitis, suggesting its potential as a therapeutic intervention. Additionally, it offers insights into the complex molecular mechanisms underlying paeoniflorin's anti-inflammatory properties in treating corticosteroid-dependent dermatitis.


Sujet(s)
Cytokines , Cellules dendritiques , Glucosides , Monoterpènes , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Glucosides/pharmacologie , Glucosides/usage thérapeutique , Animaux , Cytokines/métabolisme , Monoterpènes/pharmacologie , Monoterpènes/usage thérapeutique , Humains , Souris , Dermatite/traitement médicamenteux , Dermatite/immunologie , Dermatite/métabolisme , Interleukine-6/métabolisme , Simulation de docking moléculaire , Peau/anatomopathologie , Peau/effets des médicaments et des substances chimiques , Peau/immunologie , Peau/métabolisme , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Modèles animaux de maladie humaine , Antigène CD80/métabolisme , Antigène CD86/métabolisme , Mâle , Lymphopoïétine stromale thymique , Activation des lymphocytes/effets des médicaments et des substances chimiques
8.
AAPS J ; 26(4): 68, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38862748

RÉSUMÉ

Bispecific and multispecific agents have become increasingly utilized in cancer treatment and immunotherapy, yet their complex design parameters present a challenge in developing successful therapeutics. Bispecifics that crosslink receptors on two opposing cells can provide specific activation of a receptor only when these cells are in close spatial proximity, such as an immune cell and cancer cell in a tumor. These agents, including T cell activating bispecifics, can avoid off-tumor toxicity through activation only in the tumor microenvironment by utilizing a tumor target to cluster T-cell receptors for a selective costimulatory signal. Here, we investigate a panel of PD-1/CD137 targeted Humabody VH domains to determine the key factors for T cell activation, such as affinity, valency, expression level, domain orientation, and epitope location. Target expression is a dominant factor determining both specificity and potency of T cell activation. Given an intrinsic expression level, the affinity can be tuned to modulate the level of activation and IC50 and achieve specificity between low and high expression levels. Changing the epitope location and linker length showed minor improvements to activation at low expression levels, but increasing the valency for the target decreased activation at all expression levels. By combining non-overlapping epitopes for the target, we achieved higher receptor activation at low expression levels. A kinetic model was able to capture these trends, offering support for the mechanistic interpretation. This work provides a framework to quantify factors for T cell activation by cell-crosslinking bispecific agents and guiding principles for the design of new agents.


Sujet(s)
Anticorps bispécifiques , Activation des lymphocytes , Récepteur-1 de mort cellulaire programmée , Lymphocytes T , Antigènes CD137 , Anticorps bispécifiques/pharmacologie , Anticorps bispécifiques/immunologie , Humains , Antigènes CD137/immunologie , Lymphocytes T/immunologie , Lymphocytes T/effets des médicaments et des substances chimiques , Activation des lymphocytes/effets des médicaments et des substances chimiques , Récepteur-1 de mort cellulaire programmée/immunologie , Réactifs réticulants/composition chimique , Conception de médicament
9.
Clin Transl Sci ; 17(6): e13860, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38923308

RÉSUMÉ

The recommended immunosuppressive treatment after kidney transplantation consists of tacrolimus, mycophenolate mofetil, and low-dose corticosteroids. Drug concentrations are monitored using therapeutic drug monitoring (TDM), which does not necessarily correlate with pharmacodynamic activity. To find the balance between optimal efficacy and minimal toxicity, it might be more informative to monitor patients' immunological status rather than drug concentrations. We selected a panel of T-cell-based immune assays, which were used for immunomonitoring of 14 stable kidney transplantation patients. Whole blood was incubated with a T-cell stimulus, after which T-cell proliferation, T-cell activation marker expression and cytokine production were measured to study residual immune activity in vitro (before drug intake; drug added to the incubation) and ex vivo (after drug intake). T-cell proliferation was completely suppressed in all patients over the full day, while IL-2, IFN-γ, CD71, and CD154 showed fluctuations over the day with a strong inhibition (75%-25%) at 2 h post-dose. The level of inhibition was variable between patients and could not be related to pharmacokinetic parameters or the presence of regulatory or senescence immune cells. Moreover, the level of inhibition did not correlate with the in vitro tacrolimus drug effect as studied by incubating pre-dose blood samples with additional tacrolimus. Overall, IL-2, IFN-γ, CD71, and CD154 seem to be good markers to monitor residual immune activity of transplantation patients. To evaluate the correlation between these pharmacodynamic biomarkers and clinical outcome, prospective observational studies are needed.


Sujet(s)
Prolifération cellulaire , Surveillance des médicaments , Immunosuppresseurs , Transplantation rénale , Activation des lymphocytes , Lymphocytes T , Tacrolimus , Humains , Mâle , Immunosuppresseurs/administration et posologie , Immunosuppresseurs/pharmacocinétique , Immunosuppresseurs/usage thérapeutique , Adulte d'âge moyen , Femelle , Lymphocytes T/immunologie , Lymphocytes T/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Adulte , Tacrolimus/administration et posologie , Tacrolimus/pharmacocinétique , Tacrolimus/pharmacologie , Activation des lymphocytes/effets des médicaments et des substances chimiques , Surveillance des médicaments/méthodes , Sujet âgé , Acide mycophénolique/administration et posologie , Acide mycophénolique/pharmacocinétique , Interféron gamma/métabolisme
10.
Biomolecules ; 14(6)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38927036

RÉSUMÉ

Transient Receptor Potential Ankyrin 1 (TRPA1) is a non-selective cation channel involved in sensitivity to a plethora of irritating agents and endogenous mediators of oxidative stress. TRPA1 influences neuroinflammation and macrophage and lymphocyte functions, but its role is controversial in immune cells. We reported earlier a detectable, but orders-of-magnitude-lower level of Trpa1 mRNA in monocytes and lymphocytes than in sensory neurons by qRT-PCR analyses of cells from lymphoid organs of mice. Our present goals were to (a) further elucidate the expression of Trpa1 mRNA in immune cells by RNAscope in situ hybridization (ISH) and (b) test the role of TRPA1 in lymphocyte activation. RNAscope ISH confirmed that Trpa1 transcripts were detectable in CD14+ and CD4+ cells from the peritoneal cavity of mice. A selective TRPA1 agonist JT010 elevated Ca2+ levels in these cells only at high concentrations. However, a concentration-dependent inhibitory effect of JT010 was observed on T-cell receptor (TcR)-induced Ca2+ signals in CD4+ T lymphocytes, while JT010 neither modified B cell activation nor ionomycin-stimulated Ca2+ level. Based on our present and past findings, TRPA1 activation negatively modulates T lymphocyte activation, but it does not appear to be a key regulator of TcR-stimulated calcium signaling.


Sujet(s)
Activation des lymphocytes , Membre-1 de la sous-famille A de canaux cationiques à potentiel de récepteur transitoire , Membre-1 de la sous-famille A de canaux cationiques à potentiel de récepteur transitoire/métabolisme , Membre-1 de la sous-famille A de canaux cationiques à potentiel de récepteur transitoire/génétique , Animaux , Souris , Activation des lymphocytes/effets des médicaments et des substances chimiques , Lymphocytes T/métabolisme , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/immunologie , Ligands , Lymphocytes T CD4+/métabolisme , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Acétanilides/pharmacologie , Souris de lignée C57BL , Calcium/métabolisme , Récepteurs aux antigènes des cellules T/métabolisme , ARN messager/métabolisme , ARN messager/génétique , Mâle , Signalisation calcique/effets des médicaments et des substances chimiques
11.
Sci Rep ; 14(1): 13980, 2024 06 17.
Article de Anglais | MEDLINE | ID: mdl-38886484

RÉSUMÉ

Maraviroc (MVC) is an antiretroviral drug capable of binding to CCR5 receptors and block HIV entry into target cells. Moreover, MVC can activate NF-kB pathway and induce viral transcription in HIV-infected cells, being proposed as a latency reversal agent (LRA) in HIV cure strategies. However, the evaluation of immunological and metabolic parameters induced by MVC concentrations capable of inducing HIV transcription have not been explored in depth. We cultured isolated CD4 T cells in the absence or presence of MVC, and evaluated the frequency of CD4 T cell subpopulations and activation markers levels by flow cytometry, and the oxidative and glycolytic metabolic rates of CD4 T cells using a Seahorse Analyzer. Our results indicate that a high concentration of MVC did not increase the levels of activation markers, as well as glycolytic or oxidative metabolic rates in CD4 T cells. Furthermore, MVC did not induce significant changes in the frequency and activation levels of memory cell subpopulations. Our data support a safety profile of MVC as a promising LRA candidate since it does not induce alterations of the immunological and metabolic parameters that could affect the functionality of these immune cells.


Sujet(s)
Lymphocytes T CD4+ , Maraviroc , Maraviroc/pharmacologie , Lymphocytes T CD4+/métabolisme , Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Lymphocytes T CD4+/immunologie , Humains , Glycolyse/effets des médicaments et des substances chimiques , Infections à VIH/traitement médicamenteux , Infections à VIH/métabolisme , Infections à VIH/immunologie , Cellules cultivées , Triazoles/pharmacologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Activation des lymphocytes/effets des médicaments et des substances chimiques , Mâle , Antagonistes des récepteurs CCR5/pharmacologie , Cyclohexanes/pharmacologie , Adulte
12.
Sci Rep ; 14(1): 14595, 2024 06 25.
Article de Anglais | MEDLINE | ID: mdl-38918496

RÉSUMÉ

There are two known mechanisms by which natural killer (NK) cells recognize and kill diseased targets: (i) direct killing and (ii) antibody-dependent cell-mediated cytotoxicity (ADCC). We investigated an indirect NK cell activation strategy for the enhancement of human NK cell killing function. We did this by leveraging the fact that toll-like receptor 9 (TLR9) agonism within pools of human peripheral blood mononuclear cells (PBMCs) results in a robust interferon signaling cascade that leads to NK cell activation. After TLR9 agonist stimulation, NK cells were enriched and incorporated into assays to assess their ability to kill tumor cell line targets. Notably, differential impacts of TLR9 agonism were observed-direct killing was enhanced while ADCC was not increased. To ensure that the observed differential effects were not attributable to differences between human donors, we recapitulated the observation using our Natural Killer-Simultaneous ADCC and Direct Killing Assay (NK-SADKA) that controls for human-to-human differences. Next, we observed a treatment-induced decrease in NK cell surface CD16-known to be shed by NK cells post-activation. Given the essential role of CD16 in ADCC, such shedding could account for the observed differential impact of TLR9 agonism on NK cell-mediated killing capacity.


Sujet(s)
Cytotoxicité à médiation cellulaire dépendante des anticorps , Cellules tueuses naturelles , Récepteur-9 de type Toll-like , Humains , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/effets des médicaments et des substances chimiques , Cellules tueuses naturelles/métabolisme , Cytotoxicité à médiation cellulaire dépendante des anticorps/effets des médicaments et des substances chimiques , Récepteur-9 de type Toll-like/agonistes , Récepteur-9 de type Toll-like/métabolisme , Agranulocytes/métabolisme , Agranulocytes/immunologie , Agranulocytes/effets des médicaments et des substances chimiques , Activation des lymphocytes/effets des médicaments et des substances chimiques , Activation des lymphocytes/immunologie , Récepteurs du fragment Fc des IgG/métabolisme , Récepteurs du fragment Fc des IgG/immunologie , Lignée cellulaire tumorale , Cytotoxicité immunologique/effets des médicaments et des substances chimiques
13.
PLoS One ; 19(6): e0305261, 2024.
Article de Anglais | MEDLINE | ID: mdl-38923962

RÉSUMÉ

Inhibiting the functional role of negative regulators in immune cells is an effective approach for developing immunotherapies. The serine/threonine kinase hematopoietic progenitor kinase 1 (HPK1) involved in the T-cell receptor signaling pathway attenuates T-cell activation by inducing the degradation of SLP-76 through its phosphorylation at Ser-376, reducing the immune response. Interestingly, several studies have shown that the genetic ablation or pharmacological inhibition of HPK1 kinase activity improves the immune response to cancers by enhancing T-cell activation and cytokine production; therefore, HPK1 could be a promising druggable target for T-cell-based cancer immunotherapy. To increase the immune response against cancer cells, we designed and synthesized KHK-6 and evaluated its cellular activity to inhibit HPK1 and enhance T-cell activation. KHK-6 inhibited HPK1 kinase activity with an IC50 value of 20 nM and CD3/CD28-induced phosphorylation of SLP-76 at Ser-376 Moreover, KHK-6 significantly enhanced CD3/CD28-induced production of cytokines; proportion of CD4+ and CD8+ T cells that expressed CD69, CD25, and HLA-DR markers; and T-cell-mediated killing activity of SKOV3 and A549 cells. In conclusion, KHK-6 is a novel ATP-competitive HPK1 inhibitor that blocks the phosphorylation of HPK1 downstream of SLP-76, enhancing the functional activation of T cells. In summary, our study showed the usefulness of KHK-6 in the drug discovery for the HPK1-inhibiting immunotherapy.


Sujet(s)
Activation des lymphocytes , Protein-Serine-Threonine Kinases , Humains , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Activation des lymphocytes/effets des médicaments et des substances chimiques , Phosphorylation/effets des médicaments et des substances chimiques , Lymphocytes T/immunologie , Lymphocytes T/effets des médicaments et des substances chimiques , Protéines adaptatrices de la transduction du signal/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Cytokines/métabolisme , Phosphoprotéines/métabolisme
14.
Nat Commun ; 15(1): 5483, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38942804

RÉSUMÉ

Dexamethasone is the standard of care for critically ill patients with COVID-19, but the mechanisms by which it decreases mortality and its immunological effects in this setting are not understood. Here we perform bulk and single-cell RNA sequencing of samples from the lower respiratory tract and blood, and assess plasma cytokine profiling to study the effects of dexamethasone on both systemic and pulmonary immune cell compartments. In blood samples, dexamethasone is associated with decreased expression of genes associated with T cell activation, including TNFSFR4 and IL21R. We also identify decreased expression of several immune pathways, including major histocompatibility complex-II signaling, selectin P ligand signaling, and T cell recruitment by intercellular adhesion molecule and integrin activation, suggesting these are potential mechanisms of the therapeutic benefit of steroids in COVID-19. We identify additional compartment- and cell- specific differences in the effect of dexamethasone that are reproducible in publicly available datasets, including steroid-resistant interferon pathway expression in the respiratory tract, which may be additional therapeutic targets. In summary, we demonstrate compartment-specific effects of dexamethasone in critically ill COVID-19 patients, providing mechanistic insights with potential therapeutic relevance. Our results highlight the importance of studying compartmentalized inflammation in critically ill patients.


Sujet(s)
Traitements médicamenteux de la COVID-19 , COVID-19 , Cytokines , Dexaméthasone , Poumon , SARS-CoV-2 , Dexaméthasone/usage thérapeutique , Dexaméthasone/pharmacologie , Humains , COVID-19/immunologie , COVID-19/virologie , SARS-CoV-2/effets des médicaments et des substances chimiques , Poumon/effets des médicaments et des substances chimiques , Poumon/virologie , Cytokines/métabolisme , Cytokines/sang , Maladie grave , Mâle , Analyse sur cellule unique , Femelle , Adulte d'âge moyen , Lymphocytes T/immunologie , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/métabolisme , Sujet âgé , Activation des lymphocytes/effets des médicaments et des substances chimiques
15.
Cell Rep Med ; 5(6): 101590, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38843844

RÉSUMÉ

Despite the important breakthroughs of immune checkpoint inhibitors in recent years, the objective response rates remain limited. Here, we synthesize programmed cell death protein-1 (PD-1) antibody-iRGD cyclic peptide conjugate (αPD-1-(iRGD)2) through glycoengineering methods. In addition to enhancing tissue penetration, αPD-1-(iRGD)2 simultaneously engages tumor cells and PD-1+ T cells via dual targeting, thus mediating tumor-specific T cell activation and proliferation with mild effects on non-specific T cells. In multiple syngeneic mouse models, αPD-1-(iRGD)2 effectively reduces tumor growth with satisfactory biosafety. Moreover, results of flow cytometry and single-cell RNA-seq reveal that αPD-1-(iRGD)2 remodels the tumor microenvironment and expands a population of "better effector" CD8+ tumor infiltrating T cells expressing stem- and memory-associated genes, including Tcf7, Il7r, Lef1, and Bach2. Conclusively, αPD-1-(iRGD)2 is a promising antibody conjugate therapeutic beyond antibody-drug conjugate for cancer immunotherapy.


Sujet(s)
Récepteur-1 de mort cellulaire programmée , Microenvironnement tumoral , Animaux , Récepteur-1 de mort cellulaire programmée/immunologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Souris , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Humains , Lignée cellulaire tumorale , Lymphocytes T/immunologie , Lymphocytes T/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Oligopeptides/composition chimique , Oligopeptides/pharmacologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Immunoconjugués/pharmacologie , Immunoconjugués/composition chimique , Femelle , Activation des lymphocytes/effets des médicaments et des substances chimiques , Activation des lymphocytes/immunologie , Lymphocytes TIL/immunologie , Lymphocytes TIL/effets des médicaments et des substances chimiques , Inhibiteurs de points de contrôle immunitaires/pharmacologie
16.
Int J Mol Sci ; 25(11)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38892082

RÉSUMÉ

Mucosal-associated invariant T (MAIT) cells, a subset of Vα7.2+ T cells, are a crucial link between innate and adaptive immunity, responding to various stimuli through TCR-dependent and independent pathways. We investigated the responses of MAIT cells and Vα7.2+/CD161- T cells to different stimuli and evaluated the effects of Cyclosporin A (CsA) and Vitamin D3 (VitD). Peripheral blood mononuclear cells (PBMCs) from healthy donors were stimulated with various agents (PMA/Ionomycin, 5-OP-RU, 5-OP-RU/IL-12/IL-33) with or without CsA and VitD. Flow cytometric analysis assessed surface markers and intracellular cytokine production. Under steady-state conditions, MAIT cells displayed elevated expression of CCR6 and IL-13. They showed upregulated activation and exhaustion markers after activation, producing IFNγ, TNFα, and TNFα/GzB. CsA significantly inhibited MAIT cell activation and cytokine production. Conversely, Vα7.2+/CD161- T cells exhibited distinct responses, showing negligible responses to 5-OP-RU ligand but increased cytokine production upon PMA stimulation. Our study underscores the distinct nature of MAIT cells compared to Vα7.2+/CD161- T cells, which resemble conventional T cells. CsA emerges as a potent immunosuppressive agent, inhibiting proinflammatory cytokine production in MAIT cells. At the same time, VitD supports MAIT cell activation and IL-13 production, shedding light on potential therapeutic avenues for immune modulation.


Sujet(s)
Cellules T invariantes associées aux muqueuses , Sous-famille B des récepteurs de cellules NK de type lectine , Humains , Cellules T invariantes associées aux muqueuses/immunologie , Cellules T invariantes associées aux muqueuses/métabolisme , Cellules T invariantes associées aux muqueuses/effets des médicaments et des substances chimiques , Sous-famille B des récepteurs de cellules NK de type lectine/métabolisme , Activation des lymphocytes/effets des médicaments et des substances chimiques , Activation des lymphocytes/immunologie , Facteurs immunologiques/pharmacologie , Cytokines/métabolisme , Ciclosporine/pharmacologie , Cholécalciférol/pharmacologie , Agranulocytes/effets des médicaments et des substances chimiques , Agranulocytes/métabolisme , Agranulocytes/immunologie
17.
Front Immunol ; 15: 1362152, 2024.
Article de Anglais | MEDLINE | ID: mdl-38835768

RÉSUMÉ

Introduction: The effector function of T cells is regulated via immune checkpoints, activating or inhibiting the immune response. The BTLA-HVEM complex, the inhibitory immune checkpoint, may act as one of the tumor immune escape mechanisms. Therefore, interfering with the binding of these proteins can prove beneficial in cancer treatment. Our study focused on peptides interacting with HVEM at the same place as BTLA, thus disrupting the BTLA-HVEM interaction. These peptides' structure and amino acid sequences are based on the gD protein, the ligand of HVEM. Here, we investigated their immunomodulatory potential in melanoma patients. Methods: Flow cytometry analyses of activation, proliferation, and apoptosis of T cells from patients were performed. Additionally, we evaluated changes within the T cell memory compartment. Results: The most promising compound - Pep(2), increased the percentages of activated T cells and promoted their proliferation. Additionally, this peptide affected the proliferation rate and apoptosis of melanoma cell line in co-culture with T cells. Discussion: We conclude that the examined peptide may act as a booster for the immune system. Moreover, the adjuvant and activating properties of the gD-derived peptide could be used in a combinatory therapy with currently used ICI-based treatment. Our studies also demonstrate that even slight differences in the amino acid sequence of peptides and any changes in the position of the disulfide bond can strongly affect the immunomodulatory properties of compounds.


Sujet(s)
Activation des lymphocytes , Mélanome , Récepteurs immunologiques , Membre-14 de la superfamille des récepteurs au TNF , Lymphocytes T , Humains , Mélanome/immunologie , Mélanome/traitement médicamenteux , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/immunologie , Membre-14 de la superfamille des récepteurs au TNF/métabolisme , Membre-14 de la superfamille des récepteurs au TNF/immunologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Activation des lymphocytes/effets des médicaments et des substances chimiques , Femelle , Mâle , Adulte d'âge moyen , Prolifération cellulaire/effets des médicaments et des substances chimiques , Sujet âgé , Lignée cellulaire tumorale , Adulte , Apoptose/effets des médicaments et des substances chimiques , Peptides/pharmacologie , Peptides/immunologie , Gangliosides/immunologie
18.
Sci Rep ; 14(1): 13133, 2024 06 07.
Article de Anglais | MEDLINE | ID: mdl-38849432

RÉSUMÉ

The short-lived nature and heterogeneity of Natural Killer (NK) cells limit the development of NK cell-based therapies, despite their proven safety and efficacy against cancer. Here, we describe the biological basis, detailed phenotype and function of long-lived anti-tumour human NK cells (CD56highCD16+), obtained without cell sorting or feeder cells, after priming of peripheral blood cells with Bacillus Calmette-Guérin (BCG). Further, we demonstrate that survival doses of a cytokine combination, excluding IL18, administered just weekly to BCG-primed NK cells avoids innate lymphocyte exhaustion and leads to specific long-term proliferation of innate cells that exert potent cytotoxic function against a broad range of solid tumours, mainly through NKG2D. Strikingly, a NKG2C+CD57-FcεRIγ+ NK cell population expands after BCG and cytokine stimulation, independently of HCMV serology. This strategy was exploited to rescue anti-tumour NK cells even from the suppressor environment of cancer patients' bone marrow, demonstrating that BCG confers durable anti-tumour features to NK cells.


Sujet(s)
Prolifération cellulaire , Cellules tueuses naturelles , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/effets des médicaments et des substances chimiques , Humains , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tumeurs/immunologie , Tumeurs/traitement médicamenteux , Vaccin BCG/immunologie , Vaccin BCG/administration et posologie , Mycobacterium bovis/immunologie , Activation des lymphocytes/effets des médicaments et des substances chimiques , Sous-famille K des récepteurs de cellules NK de type lectine/métabolisme , Interleukines/métabolisme , Antigènes CD56/métabolisme , Sous-famille C des récepteurs de cellules NK de type lectine/métabolisme
19.
Molecules ; 29(12)2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38930843

RÉSUMÉ

Cyclophilin A (CypA), the cellular receptor of the immunosuppressant cyclosporin A (CsA), is an abundant cytosolic protein and is involved in a variety of diseases. For example, CypA supports cancer proliferation and mediates viral infections, such as the human immunodeficiency virus 1 (HIV-1). Here, we present the design of PROTAC (proteolysis targeting chimera) compounds against CypA to induce its intracellular proteolysis and to investigate their effect on immune cells. Interestingly, upon connecting to E3 ligase ligands, both peptide-based low-affinity binders and CsA-based high-affinity binders can degrade CypA at nM concentration in HeLa cells and fibroblast cells. As the immunosuppressive effect of CsA is not directly associated with the binding of CsA to CypA but the inhibition of phosphatase calcineurin by the CypA:CsA complex, we investigated whether a CsA-based PROTAC compound could induce CypA degradation without affecting the activation of immune cells. P3, the most efficient PROTAC compound discovered from this study, could deplete CypA in lymphocytes without affecting cell proliferation and cytokine production. This work demonstrates the feasibility of the PROTAC approach in depleting the abundant cellular protein CypA at low drug dosage without affecting immune cells, allowing us to investigate the potential therapeutic effects associated with the endogenous protein in the future.


Sujet(s)
Cyclophiline A , Ciclosporine , Activation des lymphocytes , Protéolyse , Lymphocytes T , Humains , Cyclophiline A/métabolisme , Ciclosporine/pharmacologie , Protéolyse/effets des médicaments et des substances chimiques , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/métabolisme , Activation des lymphocytes/effets des médicaments et des substances chimiques , Cellules HeLa , Prolifération cellulaire/effets des médicaments et des substances chimiques , Immunosuppresseurs/pharmacologie , Immunosuppresseurs/composition chimique , Chimère ciblant la protéolyse
20.
Molecules ; 29(12)2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38930852

RÉSUMÉ

Nutraceutical immune support offers potential for designing blends with complementary mechanisms of action for robust support of innate immune alertness. We documented enhanced immune activation when bovine colostrum peptides (BC-Pep) were added to an immune blend (IB) containing ß-glucans from yeast, shiitake, maitake, and botanical non-ß-glucan polysaccharides. Human peripheral blood mononuclear cells (PBMCs) were cultured with IB, BC-Pep, and IB + BC-Pep for 20 h, whereafter expression of the activation marker CD69 was evaluated on NK cells, NKT cells, and T cells. Cytokine levels were tested in culture supernatants. PBMCs were co-cultured with K562 target cells to evaluate T cell-mediated cytotoxicity. IB + BC-Pep triggered highly significant increases in IL-1ß, IL-6, and TNF-α, above that of cultures treated with matching doses of either IB or BC-Pep. NK cell and T cell activation was increased by IB + BC-Pep, reaching levels of CD69 expression several fold higher than either BC-Pep or IB alone. IB + BC-Pep significantly increased T cell-mediated cytotoxic killing of K562 target cells. This synergistic effect suggests unique amplification of signal transduction of NK cells and T cells due to modulation of IB-induced signaling pathways by BC-Pep and is of interest for further pre-clinical and clinical testing of immune defense activity against virally infected and transformed cells.


Sujet(s)
Colostrum , Immunité innée , Peptides , bêta-Glucanes , Animaux , Bovins , Humains , Colostrum/composition chimique , Colostrum/immunologie , Immunité innée/effets des médicaments et des substances chimiques , bêta-Glucanes/pharmacologie , bêta-Glucanes/composition chimique , Peptides/pharmacologie , Peptides/composition chimique , Cellules tueuses naturelles/effets des médicaments et des substances chimiques , Cellules tueuses naturelles/immunologie , Cytokines/métabolisme , Activation des lymphocytes/effets des médicaments et des substances chimiques , Agranulocytes/effets des médicaments et des substances chimiques , Agranulocytes/immunologie , Agranulocytes/métabolisme , Agaricales/composition chimique , Antigènes de différenciation des lymphocytes T/métabolisme , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Cellules K562 , Antigènes CD/métabolisme , Lectines de type C
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE