Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.057
Filtrer
1.
Front Immunol ; 15: 1415102, 2024.
Article de Anglais | MEDLINE | ID: mdl-39007132

RÉSUMÉ

Human regulatory T cells (Treg) suppress other immune cells. Their dysfunction contributes to the pathophysiology of autoimmune diseases, including type 1 diabetes (T1D). Infusion of Tregs is being clinically evaluated as a novel way to prevent or treat T1D. Genetic modification of Tregs, most notably through the introduction of a chimeric antigen receptor (CAR) targeting Tregs to pancreatic islets, may improve their efficacy. We evaluated CAR targeting of human Tregs to monocytes, a human ß cell line and human islet ß cells in vitro. Targeting of HLA-A2-CAR (A2-CAR) bulk Tregs to HLA-A2+ cells resulted in dichotomous cytotoxic killing of human monocytes and islet ß cells. In exploring subsets and mechanisms that may explain this pattern, we found that CD39 expression segregated CAR Treg cytotoxicity. CAR Tregs from individuals with more CD39low/- Tregs and from individuals with genetic polymorphism associated with lower CD39 expression (rs10748643) had more cytotoxicity. Isolated CD39- CAR Tregs had elevated granzyme B expression and cytotoxicity compared to the CD39+ CAR Treg subset. Genetic overexpression of CD39 in CD39low CAR Tregs reduced their cytotoxicity. Importantly, ß cells upregulated protein surface expression of PD-L1 and PD-L2 in response to A2-CAR Tregs. Blockade of PD-L1/PD-L2 increased ß cell death in A2-CAR Treg co-cultures suggesting that the PD-1/PD-L1 pathway is important in protecting islet ß cells in the setting of CAR immunotherapy. In summary, introduction of CAR can enhance biological differences in subsets of Tregs. CD39+ Tregs represent a safer choice for CAR Treg therapies targeting tissues for tolerance induction.


Sujet(s)
Apyrase , Récepteurs chimériques pour l'antigène , Lymphocytes T régulateurs , Humains , Apyrase/immunologie , Apyrase/métabolisme , Lymphocytes T régulateurs/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/métabolisme , Cytotoxicité immunologique , Ilots pancréatiques/immunologie , Ilots pancréatiques/métabolisme , Diabète de type 1/immunologie , Diabète de type 1/thérapie , Antigène HLA-A2/immunologie , Antigène HLA-A2/génétique , Antigène HLA-A2/métabolisme , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Cellules à insuline/immunologie , Cellules à insuline/métabolisme , Antigènes CD
2.
Front Immunol ; 15: 1397967, 2024.
Article de Anglais | MEDLINE | ID: mdl-38947317

RÉSUMÉ

Introduction: CD39 plays an important role in the immunoregulation and inhibition of effector cells. It is expressed on immune cells, including Tregs, and on extracellular vesicles (EVs) budding from the plasma membrane. Platelet transfusion may induce alloimmunization against HLA-I antigens, leading to refractoriness to platelet transfusion with severe consequences for patients. Tregs may play a key role in determining whether alloimmunization occurs in patients with hematologic disorders. We hypothesized that CD39+ EVs might play an immunoregulatory role, particularly in the context of platelet transfusions in patients with hematologic disorders. Such alloimmunization leads to the production of alloantibodies and is sensitive to the regulatory action of CD39. Methods: We characterized CD39+ EVs in platelet concentrates by flow cytometry. The absolute numbers and cellular origins of CD39+ EVs were evaluated. We also performed functional tests to evaluate interactions with immune cells and their functions. Results: We found that CD39+ EVs from platelet concentrates had an inhibitory phenotype that could be transferred to the immune cells with which they interacted: CD4+ and CD8+ T lymphocytes (TLs), dendritic cells, monocytes, and B lymphocytes (BLs). Moreover, the concentration of CD39+ EVs in platelet concentrates varied and was very high in 10% of concentrates. The number of these EVs present was determinant for EV-cell interactions. Finally, functional interactions were observed with BLs, CD4+ TLs and CD39+ EVs for immunoglobulin production and lymphoproliferation, with potential implications for the immunological management of patients.


Sujet(s)
Plaquettes , Vésicules extracellulaires , Antigène CD9 , Humains , Vésicules extracellulaires/immunologie , Vésicules extracellulaires/métabolisme , Plaquettes/immunologie , Plaquettes/métabolisme , Antigène CD9/métabolisme , Communication cellulaire/immunologie , Transfusion de plaquettes , Femelle , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Mâle , Apyrase/métabolisme , Apyrase/immunologie , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Antigènes CD
3.
Nat Commun ; 15(1): 5759, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38982116

RÉSUMÉ

Type I interferons have been well recognized for their roles in various types of immune cells during tumor immunotherapy. However, their direct effects on tumor cells are less understood. Oxidative phosphorylation is typically latent in tumor cells. Whether oxidative phosphorylation can be targeted for immunotherapy remains unclear. Here, we find that tumor cell responsiveness to type I, but not type II interferons, is essential for CD47-SIRPα blockade immunotherapy in female mice. Mechanistically, type I interferons directly reprogram tumor cell metabolism by activating oxidative phosphorylation for ATP production in an ISG15-dependent manner. ATP extracellular release is also promoted by type I interferons due to enhanced secretory autophagy. Functionally, tumor cells with genetic deficiency in oxidative phosphorylation or autophagy are resistant to CD47-SIRPα blockade. ATP released upon CD47-SIRPα blockade is required for antitumor T cell response induction via P2X7 receptor-mediated dendritic cell activation. Based on this mechanism, combinations with inhibitors of ATP-degrading ectoenzymes, CD39 and CD73, are designed and show synergistic antitumor effects with CD47-SIRPα blockade. Together, these data reveal an important role of type I interferons on tumor cell metabolic reprograming for tumor immunotherapy and provide rational strategies harnessing this mechanism for enhanced efficacy of CD47-SIRPα blockade.


Sujet(s)
Adénosine triphosphate , Antigènes CD47 , Interféron de type I , Phosphorylation oxydative , Récepteurs immunologiques , Transduction du signal , Animaux , Antigènes CD47/métabolisme , Antigènes CD47/génétique , Interféron de type I/métabolisme , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/génétique , Femelle , Souris , Adénosine triphosphate/métabolisme , Phosphorylation oxydative/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Souris de lignée C57BL , Immunothérapie/méthodes , Humains , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Récepteurs purinergiques P2X7/métabolisme , Récepteurs purinergiques P2X7/génétique , Autophagie/effets des médicaments et des substances chimiques , Apyrase/métabolisme , Souris knockout , Tumeurs/immunologie , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Cytokines/métabolisme
4.
Cell Rep Med ; 5(7): 101639, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38959887

RÉSUMÉ

Environmental enteric dysfunction (EED) is a condition associated with malnutrition that can progress to malabsorption and villous atrophy. Severe EED results in linear growth stunting, slowed neurocognitive development, and unresponsiveness to oral vaccines. Prenatal exposure to malnutrition and breast feeding by malnourished mothers replicates EED. Pups are characterized by deprivation of secretory IgA (SIgA) and altered development of the gut immune system and microbiota. Extracellular ATP (eATP) released by microbiota limits T follicular helper (Tfh) cell activity and SIgA generation in Peyer's patches (PPs). Administration of a live biotherapeutic releasing the ATP-degrading enzyme apyrase to malnourished pups restores SIgA levels and ameliorates stunted growth. SIgA is instrumental in improving the growth and intestinal immune competence of mice while they are continuously fed a malnourished diet. The analysis of microbiota composition suggests that amplification of endogenous SIgA may exert a dominant function in correcting malnourishment dysbiosis and its consequences on host organisms, irrespective of the actual microbial ecology.


Sujet(s)
Microbiome gastro-intestinal , Immunoglobuline A sécrétoire , Malnutrition , Animaux , Immunoglobuline A sécrétoire/métabolisme , Malnutrition/immunologie , Souris , Femelle , Animaux nouveau-nés , Humains , Apyrase/métabolisme , Nouveau-né
5.
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
6.
Oncoimmunology ; 13(1): 2371051, 2024.
Article de Anglais | MEDLINE | ID: mdl-38915783

RÉSUMÉ

Improving cancer immunotherapy efficacy hinges on identifying key T-cell populations critical for tumor control and response to Immune Checkpoint Blockade (ICB). We have recently reported that while the co-expression of PD-1 and CD28 is associated with impaired functionality in peripheral blood, it significantly enhances T-cell fitness in the tumor site of non-small cell lung cancer (NSCLC) patients. To uncover the underlying mechanisms, we explored the role of CD26, a key player in T-cell activation through its interaction with adenosine deaminase (ADA), a crucial intra/extracellular enzyme able to neutralize local adenosine (ADO). We found that an autocrine ADA/CD26 axis enhances CD8+PD-1+CD28+ T-cell function, particularly within an immunosuppressive environment marked by CD39 expression. Then, we interrogated the TCGA and OAK datasets to gain insight into the prognostic/predictive potential of our findings. We identified a signature predicting overall survival (OS) in LUAD patients and response to atezolizumab in advanced LUAD cases. These findings suggest promising avenues for therapeutic intervention targeting the ADA/CD26 axis.


Sujet(s)
Adenosine deaminase , Antigène CD28 , Lymphocytes T CD8+ , Carcinome pulmonaire non à petites cellules , Dipeptidyl peptidase 4 , Inhibiteurs de points de contrôle immunitaires , Tumeurs du poumon , Récepteur-1 de mort cellulaire programmée , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/anatomopathologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Antigène CD28/métabolisme , Pronostic , Récepteur-1 de mort cellulaire programmée/métabolisme , Dipeptidyl peptidase 4/métabolisme , Dipeptidyl peptidase 4/génétique , Adenosine deaminase/métabolisme , Adenosine deaminase/génétique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/pharmacologie , Anticorps monoclonaux humanisés/administration et posologie , Femelle , Mâle , Apyrase/métabolisme
8.
J Nanobiotechnology ; 22(1): 364, 2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38915007

RÉSUMÉ

Photothermal therapy (PTT) is a promising cancer treatment method due to its ability to induce tumor-specific T cell responses and enhance therapeutic outcomes. However, incomplete PTT can leave residual tumors that often lead to new metastases and decreased patient survival in clinical scenarios. This is primarily due to the release of ATP, a damage-associated molecular pattern that quickly transforms into the immunosuppressive metabolite adenosine by CD39, prevalent in the tumor microenvironment, thus promoting tumor immune evasion. This study presents a photothermal nanomedicine fabricated by electrostatic adsorption among the Fe-doped polydiaminopyridine (Fe-PDAP), indocyanine green (ICG), and CD39 inhibitor sodium polyoxotungstate (POM-1). The constructed Fe-PDAP@ICG@POM-1 (FIP) can induce tumor PTT and immunogenic cell death when exposed to a near-infrared laser. Significantly, it can inhibit the ATP-adenosine pathway by dual-directional immunometabolic regulation, resulting in increased ATP levels and decreased adenosine synthesis, which ultimately reverses the immunosuppressive microenvironment and increases the susceptibility of immune checkpoint blockade (aPD-1) therapy. With the aid of aPD-1, the dual-directional immunometabolic regulation strategy mediated by FIP can effectively suppress/eradicate primary and distant tumors and evoke long-term solid immunological memory. This study presents an immunometabolic control strategy to offer a salvage option for treating residual tumors following incomplete PTT.


Sujet(s)
Immunothérapie , Nanomédecine , Thérapie photothermique , Microenvironnement tumoral , Animaux , Thérapie photothermique/méthodes , Immunothérapie/méthodes , Souris , Nanomédecine/méthodes , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Humains , Vert indocyanine/composition chimique , Vert indocyanine/pharmacologie , Tumeurs/thérapie , Adénosine triphosphate/métabolisme , Adénosine/pharmacologie , Adénosine/composition chimique , Souris de lignée C57BL , Apyrase/métabolisme , Femelle , Photothérapie/méthodes
9.
Biochim Biophys Acta Mol Basis Dis ; 1870(6): 167219, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38734321

RÉSUMÉ

Chronic infections induce CD4+ T-cells with cytotoxic functions (CD4 CTLs); at present, it is still unknown whether latent tuberculosis (LTB) and active tuberculosis (ATB) induce CD4 CTLs. Plasma and cells from four patient groups-uninfected contact (UC), LTB, and ATB (divided as sensitive [DS-TB]- or resistant [DR-TB]-drug)-were evaluated by flow cytometry, q-PCR, and proteomics. The data showed that ATB patients had an increased frequency of CD4+ T-cells and a decreased frequency of CD8+ T-cells. The latter displays an exhausted-like profile characterized by CD39, CD279, and TIM-3 expression. ATB had a high frequency of CD4 + perforin+ cells, suggesting a CD4 CTL profile. The expression (at the transcriptional level) of granzyme A, granzyme B, granulysin, and perforin, as well as the genes T-bet (Tbx21) and NKG2D (Klrk1), in enriched CD4+ T-cells, confirmed the cytotoxic signature of CD4+ T-cells during ATB (which was stronger in DS-TB than in DR-TB). Moreover, proteomic analysis revealed the presence of HSP70 (in DS-TB) and annexin A5 (in DR-TB), which are molecules that have been associated with favoring the CD4 CTL profile. Finally, we found that lipids from Mycobacterium tuberculosis increased the presence of CD4 CTLs in DR-TB patients. Our data suggest that ATB is characterized by exhausted-like CD8+ T-cells, which, together with a specific microenvironment, favor the presence of CD4 CTLs.


Sujet(s)
Lymphocytes T CD4+ , Lymphocytes T CD8+ , Granzymes , Récepteur cellulaire-2 du virus de l'hépatite A , Perforine , Tuberculose , Humains , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Mâle , Granzymes/métabolisme , Granzymes/génétique , Granzymes/immunologie , Perforine/métabolisme , Perforine/génétique , Perforine/immunologie , Adulte , Femelle , Récepteur cellulaire-2 du virus de l'hépatite A/métabolisme , Récepteur cellulaire-2 du virus de l'hépatite A/immunologie , Tuberculose/immunologie , Tuberculose/microbiologie , Tuberculose latente/immunologie , Tuberculose latente/microbiologie , Adulte d'âge moyen , Lymphocytes T cytotoxiques/immunologie , Lymphocytes T cytotoxiques/métabolisme , Mycobacterium tuberculosis/immunologie , Protéines à domaine boîte-T/métabolisme , Protéines à domaine boîte-T/génétique , Protéines à domaine boîte-T/immunologie , Antigènes CD/métabolisme , Antigènes CD/immunologie , Antigènes CD/génétique , Sous-famille K des récepteurs de cellules NK de type lectine/métabolisme , Sous-famille K des récepteurs de cellules NK de type lectine/immunologie , Sous-famille K des récepteurs de cellules NK de type lectine/génétique , Protéomique/méthodes , Antigènes de différenciation des lymphocytes T , Apyrase
10.
Clin Immunol ; 264: 110260, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38788885

RÉSUMÉ

Sjögren's disease (SjD) is a chronic autoimmune disease characterized by focal lymphocytic inflammation in lacrimal and salivary glands. We recently identified IL-27 as a requisite signal for the spontaneous SjD-like manifestations in nonobese diabetic (NOD) mice. Here, we define T cell-intrinsic effects of IL-27 in lacrimal gland disease in NOD mice. IL-27 receptor was required by both CD4 T effector (Te) cells and CD8 T cells to mediate focal inflammation. Intrinsic IL-27 signaling was associated with PD-1 and ICOS expressing T follicular helper (Tfh)-like CD4 Te cells within lacrimal glands, including subsets defined by CD73 or CD39 expression. CD8 T cells capable of IL-27 signaling also expressed PD-1 with subsets expressing ICOS and CD73 demonstrating a T follicular cytotoxic (Tfc)-like cell phenotype and others expressing a CD39hi exhausted-like phenotype. These findings suggest IL-27 is a key early signal driving a follicular-type response in lacrimal gland inflammation in NOD mice.


Sujet(s)
Lymphocytes T CD8+ , Modèles animaux de maladie humaine , Appareil lacrymal , Souris de lignée NOD , Syndrome de Gougerot-Sjögren , Animaux , Syndrome de Gougerot-Sjögren/immunologie , Souris , Lymphocytes T CD8+/immunologie , Appareil lacrymal/immunologie , Appareil lacrymal/anatomopathologie , Interleukines/immunologie , Interleukines/métabolisme , Lymphocytes T CD4+/immunologie , Récepteur-1 de mort cellulaire programmée/immunologie , Récepteur-1 de mort cellulaire programmée/métabolisme , Femelle , Transduction du signal/immunologie , Récepteurs aux interleukines/immunologie , Interleukine-27/métabolisme , Interleukine-27/immunologie , Protéine inductible de costimulation du lymphocyte T/immunologie , Protéine inductible de costimulation du lymphocyte T/métabolisme , Apyrase/immunologie , Apyrase/métabolisme
11.
J Thromb Haemost ; 22(8): 2331-2344, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38754782

RÉSUMÉ

BACKGROUND: Ischemic stroke is characterized by a necrotic lesion in the brain surrounded by an area of dying cells termed the penumbra. Salvaging the penumbra either with thrombolysis or mechanical retrieval is the cornerstone of stroke management. At-risk neuronal cells release extracellular adenosine triphosphate, triggering microglial activation and causing a thromboinflammatory response, culminating in endothelial activation and vascular disruption. This is further aggravated by ischemia-reperfusion injury that follows all reperfusion therapies. The ecto-enzyme CD39 regulates extracellular adenosine triphosphate by hydrolyzing it to adenosine, which has antithrombotic and anti-inflammatory properties and reverses ischemia-reperfusion injury. OBJECTIVES: The objective off the study was to determine the efficacy of our therapeutic, anti-VCAM-CD39 in ischaemic stroke. METHODS: We developed anti-VCAM-CD39 that targets the antithrombotic and anti-inflammatory properties of recombinant CD39 to the activated endothelium of the penumbra by binding to vascular cell adhesion molecule (VCAM)-1. Mice were subjected to 30 minutes of middle cerebral artery occlusion and analyzed at 24 hours. Anti-VCAM-CD39 or control agents (saline, nontargeted CD39, or anti-VCAM-inactive CD39) were given at 3 hours after middle cerebral artery occlusion. RESULTS: Anti-VCAM-CD39 treatment reduced neurologic deficit; magnetic resonance imaging confirmed significantly smaller infarcts together with an increase in cerebrovascular perfusion. Anti-VCAM-CD39 also restored blood-brain barrier integrity and reduced microglial activation. Coadministration of anti-VCAM-CD39 with thrombolytics (tissue plasminogen activator [tPA]) further reduced infarct volumes and attenuated blood-brain barrier permeability with no associated increase in intracranial hemorrhage. CONCLUSION: Anti-VCAM-CD39, uniquely targeted to endothelial cells, could be a new stroke therapy even when administered 3 hours postischemia and may further synergize with thrombolytic therapy to improve stroke outcomes.


Sujet(s)
Antigènes CD , Apyrase , Barrière hémato-encéphalique , Modèles animaux de maladie humaine , Infarctus du territoire de l'artère cérébrale moyenne , Accident vasculaire cérébral ischémique , Souris de lignée C57BL , Molécule-1 d'adhérence des cellules vasculaires , Animaux , Apyrase/métabolisme , Accident vasculaire cérébral ischémique/traitement médicamenteux , Accident vasculaire cérébral ischémique/métabolisme , Infarctus du territoire de l'artère cérébrale moyenne/traitement médicamenteux , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Molécule-1 d'adhérence des cellules vasculaires/métabolisme , Antigènes CD/métabolisme , Mâle , Cellules endothéliales/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Humains , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Souris , Fibrinolytiques/usage thérapeutique , Fibrinolytiques/pharmacologie , Encéphale/métabolisme , Encéphale/anatomopathologie , Protéines recombinantes/usage thérapeutique , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/métabolisme
12.
Clinics (Sao Paulo) ; 79: 100390, 2024.
Article de Anglais | MEDLINE | ID: mdl-38781760

RÉSUMÉ

Endometriosis's pathophysiology remains incompletely understood, with evidence pointing towards a dysregulated immune response. Regulatory T (Treg) cells, pivotal in maintaining self-tolerance, may facilitate the survival of ectopic endometrial cells within the abdominal cavity, thereby contributing to endometriosis development. This study aimed to assess the prevalence of CD39+CD73+ suppressor Treg cell subsets in the peripheral blood of endometriosis patients. This research focuses on the pivotal role of regulatory T-cells (Tregs), which are essential for maintaining immune tolerance and preventing autoimmune diseases. A case-control study was conducted, including 32 women diagnosed with endometriosis and 22 control subjects. The frequency of peripheral blood CD39+CD73+ suppressor Treg cells was quantified using flow cytometry. No significant differences were observed in the frequency of CD3+CD4+CD25High cells (Median [M]: 10.1; Interquartile Range [IQR]: 6.32‒18.3 vs. M: 9.72; IQR: 6.22-19.8) or CD3+CD4+CD25HighCD39+Foxp3+ cells (M: 31.1; IQR: 19.7-44.0 vs. M: 30.55; IQR: 18.5-45.5) between controls and patients. However, a significantly lower frequency of CD3+CD4+CD25HighCD39+CD73+ cells was observed in the endometriosis group compared to controls (M: 1.98; IQR: 0.0377-3.17 vs. M: 2.25; IQR: 0.50-4.08; p = 0.0483), suggesting a reduction in systemic immune tolerance among these patients. This finding highlights the potential role of CD39 and CD73 expression on Treg cells as biomarkers for assessing disease severity and progression. Furthermore, elucidating the mechanisms driving these alterations may unveil new therapeutic strategies to restore immune equilibrium and mitigate endometriosis symptoms.


Sujet(s)
Apyrase , Endométriose , Cytométrie en flux , Facteurs de transcription Forkhead , Lymphocytes T régulateurs , Humains , Femelle , Endométriose/immunologie , Endométriose/sang , Lymphocytes T régulateurs/immunologie , Adulte , Études cas-témoins , Facteurs de transcription Forkhead/sang , Facteurs de transcription Forkhead/analyse , Apyrase/analyse , 5'-Nucleotidase/sang , Jeune adulte , Antigènes CD/sang , Antigènes CD/analyse , Statistique non paramétrique , Valeurs de référence
13.
J Am Heart Assoc ; 13(11): e033985, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38804212

RÉSUMÉ

BACKGROUND: ADP and ATP are importantly involved in vascular and thrombotic homeostasis, via multiple receptor pathways. Blockade of ADP P2Y12 receptors inhibits platelet aggregation and represents an effective cardiovascular disease prevention strategy. AZD3366 (APT102), a long-acting recombinant form of an optimized CD39L3 human apyrase, has effectively reduced ATP, ADP, and platelet aggregation and provided tissue protection in preclinical models, features that could be very beneficial in treating patients with cardiovascular disease. METHODS AND RESULTS: We conducted this phase 1, first-in-human study of single ascending doses of intravenous AZD3366 or placebo, including doses added to dual antiplatelet therapy with ticagrelor and acetylsalicylic acid. The primary objective was safety and tolerability; secondary and exploratory objectives included pharmacokinetics, pharmacodynamics (measured as inhibition of platelet aggregation), adenosine diphosphatase (ADPase) activity, and ATP/ADP metabolism. In total, 104 participants were randomized. AZD3366 was generally well tolerated, with no major safety concerns observed. ADPase activity increased in a dose-dependent manner with a strong correlation to AZD3366 exposure. Inhibition of ADP-stimulated platelet aggregation was immediate, substantial, and durable. In addition, there was a prompt decrease in systemic ATP concentration and an increase in adenosine monophosphate concentrations, whereas ADP concentration appeared generally unaltered. At higher doses, there was a prolongation of capillary bleeding time without detectable changes in the ex vivo thromboelastometric parameters. CONCLUSIONS: AZD3366 was well tolerated in healthy participants and demonstrated substantial and durable inhibition of platelet aggregation after single dosing. Higher doses prolonged capillary bleeding time without detectable changes in ex vivo thromboelastometric parameters. REGISTRATION: URL: https://www.clinicaltrials.gov; Unique Identifier: NCT04588727.


Sujet(s)
Apyrase , Acide acétylsalicylique , Antiagrégants plaquettaires , Agrégation plaquettaire , Ticagrélor , Humains , Mâle , Ticagrélor/pharmacocinétique , Ticagrélor/administration et posologie , Ticagrélor/effets indésirables , Femelle , Apyrase/métabolisme , Apyrase/administration et posologie , Agrégation plaquettaire/effets des médicaments et des substances chimiques , Acide acétylsalicylique/administration et posologie , Acide acétylsalicylique/pharmacocinétique , Acide acétylsalicylique/effets indésirables , Antiagrégants plaquettaires/pharmacocinétique , Antiagrégants plaquettaires/administration et posologie , Antiagrégants plaquettaires/effets indésirables , Adulte d'âge moyen , Adulte , Méthode en double aveugle , Bithérapie antiplaquettaire , Association de médicaments , Jeune adulte , ADP , Plaquettes/effets des médicaments et des substances chimiques , Plaquettes/métabolisme , Relation dose-effet des médicaments , Résultat thérapeutique , Protéines recombinantes/administration et posologie , Protéines recombinantes/pharmacocinétique , Antagonistes des récepteurs purinergiques P2Y/pharmacocinétique , Antagonistes des récepteurs purinergiques P2Y/administration et posologie , Antagonistes des récepteurs purinergiques P2Y/effets indésirables , Antagonistes des récepteurs purinergiques P2Y/pharmacologie
14.
Viruses ; 16(5)2024 05 20.
Article de Anglais | MEDLINE | ID: mdl-38793691

RÉSUMÉ

In patients with COVID-19, broad panels of immune checkpoint molecules (ICPMs) and the purinergic signaling have not been studied in parallel. We aimed to perform in-depth immunophenotyping of major cell subsets present in human peripheral blood of COVID-19 patients and controls using PD1, TIM3, LAG3, TIGIT, and CD200R, as well as CD39, as markers for the purinergic signaling pathway. We studied 76 COVID-19 patients and 12 healthy controls using peripheral blood mononuclear cells on flow cytometry. Univariable and multivariable statistics were performed. All ICPMs studied were significantly overexpressed on different cell subsets of COVID-19 patients when compared with healthy controls. Elevated lactate dehydrogenase; C-reactive protein; age; and high expression of CD45+, CD39+CD45+, TIM3+CD39+CD4+CD45+, and TIM3+CD39+CD8+CD3+CD4+ cells were significantly associated with severe COVID-19. On multivariable analysis, however, only high expression of CD39+CD45+ (OR 51.4, 95% CI 1.5 to 1763) and TIM3+CD39+CD4+CD3+CD45+ (OR 22.6, 95% CI 1.8 to 277) cells was an independent predictor for severe COVID-19. In conclusion, numerous ICPMs are overexpressed in COVID-19 patients when compared with healthy controls, suggesting a pathophysiological role of these molecules in SARS-CoV-2 infection. However, only TIM3 in co-expression with CD39 remained as a significant independent prognostic ICPM on multivariable analysis. The flow cytometric evaluation of TIM3+CD39+CD4+CD3+CD45+, as well as CD39+CD45+, is a powerful tool for the prognostication of COVID-19 patients on hospital admission.


Sujet(s)
Apyrase , COVID-19 , SARS-CoV-2 , Humains , COVID-19/mortalité , COVID-19/immunologie , COVID-19/diagnostic , COVID-19/sang , Mâle , Femelle , Adulte d'âge moyen , Pronostic , Sujet âgé , Études prospectives , SARS-CoV-2/immunologie , Adulte , Indice de gravité de la maladie , Protéines de points de contrôle immunitaires/génétique , Protéines de points de contrôle immunitaires/métabolisme , Antigènes CD/sang , Agranulocytes/immunologie , Immunophénotypage , Cytométrie en flux , Sujet âgé de 80 ans ou plus
15.
Sci Rep ; 14(1): 11609, 2024 05 21.
Article de Anglais | MEDLINE | ID: mdl-38773214

RÉSUMÉ

No biomarker has yet been identified that allows accurate diagnosis and prognosis of oral cancers. In this study, we investigated the presence of key metabolites in oral cancer using proton nuclear magnetic resonance (NMR) spectroscopy to identify metabolic biomarkers of gingivobuccal oral squamous cell carcinoma (GB-OSCC). NMR spectroscopy revealed that uracil was expressed in 83.09% of tumor tissues and pyrimidine metabolism was active in GB-OSCC; these results correlated well with immunohistochemistry (IHC) and RNA sequencing data. Based on further gene and protein analyses, we proposed a pathway for the production of uracil in GB-OSCC tissues. Uridinetriphosphate (UTP) is hydrolyzed to uridine diphosphate (UDP) by CD39 in the tumor microenvironment (TME). We hypothesized that UDP enters the cell with the help of the UDP-specific P2Y6 receptor for further processing by ENTPD4/5 to produce uracil. As the ATP reserves diminish, the weakened immune cells in the TME utilize pyrimidine metabolism as fuel for antitumor activity, and the same mechanism is hijacked by the tumor cells to promote their survival. Correspondingly, the differential expression of ENTPD4 and ENTPD5 in immune and tumor cells, respectively, indicatedtheir involvement in disease progression. Furthermore, higher uracil levels were detected in patients with lymph node metastasis, indicating that metastatic potential is increased in the presence of uracil. The presence of uracil and/or expression patterns of intermediate molecules in purine and pyrimidine pathways, such asCD39, CD73, and P2Y6 receptors together with ENTPD4 and ENTPD5, hold promise as biomarker(s) for oral cancer diagnosis and prognosis.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs de la bouche , Pyrimidines , Uracile , Humains , Tumeurs de la bouche/métabolisme , Tumeurs de la bouche/anatomopathologie , Uracile/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Pyrimidines/métabolisme , Femelle , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/anatomopathologie , Mâle , Adulte d'âge moyen , Microenvironnement tumoral , Sujet âgé , Apyrase/métabolisme
16.
Oncoimmunology ; 13(1): 2346359, 2024.
Article de Anglais | MEDLINE | ID: mdl-38737794

RÉSUMÉ

Immune exhaustion is a hallmark of ovarian cancer. Using multiparametric flow cytometry, the study aimed to analyze protein expression of novel immunological targets on CD3+ T cells isolated from the peripheral blood (n = 20), malignant ascites (n = 16), and tumor tissue (n = 6) of patients with ovarian cancer (OVCA). The study revealed an increased proportion of effector memory CD8+ T cells in OVCA tissue and malignant ascites. An OVCA-characteristic PD-1high CD8+ T cell population was detected, which differed from PD-1lowCD8+ T cells by increased co-expression of TIGIT, CD39, and HLA-DR. In addition, these OVCA-characteristic CD8+ T cells showed reduced expression of the transcription factor TCF-1, which may also indicate reduced effector function and memory formation. On the contrary, the transcription factor TOX, which significantly regulates terminal T cell-exhaustion, was found more frequently in these cells. Further protein and gene analysis showed that CD39 and CD73 were also expressed on OVCA tumor cells isolated from solid tumors (n = 14) and malignant ascites (n = 9). In the latter compartment, CD39 and CD73 were also associated with the expression of the "don't eat me" molecule CD24 on tumor cells. Additionally, ascites-derived CD24+EpCAM+ tumor cells showed a higher frequency of CD39+ or CD73+ cells. Furthermore, CD39 expression was associated with unfavorable clinical parameters. Expression of CD39 on T cells was upregulated through CD3/CD28 stimulation and its blockade by a newly developed nanobody construct resulted in increased proliferation (eFluor), activation (CD25 and CD134), and production of cytotoxic cytokines (IFN-γ, TNF-α, and granzyme-B) of CD8+ T cells.


Sujet(s)
Apyrase , Lymphocytes T CD8+ , Tumeurs de l'ovaire , Humains , Femelle , Tumeurs de l'ovaire/immunologie , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/métabolisme , Apyrase/métabolisme , Apyrase/génétique , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Adulte d'âge moyen , Ascites/immunologie , Ascites/anatomopathologie , Ascites/métabolisme , Antigènes CD/métabolisme , Antigènes CD/génétique , Sujet âgé , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/génétique , Récepteurs immunologiques/antagonistes et inhibiteurs , Facteur de transcription TCF-1/métabolisme , Facteur de transcription TCF-1/génétique , Antigènes HLA-DR/métabolisme , Adulte , Épuisement des cellules T , Protéines HMG
17.
Cell Mol Life Sci ; 81(1): 231, 2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38780647

RÉSUMÉ

CD200 is an anti-inflammatory protein that facilitates signal transduction through its receptor, CD200R, in cells, resulting in immune response suppression. This includes reducing M1-like macrophages, enhancing M2-like macrophages, inhibiting NK cell cytotoxicity, and downregulating CTL responses. Activation of CD200R has been found to modulate dendritic cells, leading to the induction or enhancement of Treg cells expressing Foxp3. However, the precise mechanisms behind this process are still unclear. Our previous study demonstrated that B cells in Peyer's patches can induce Treg cells, so-called Treg-of-B (P) cells, through STAT6 phosphorylation. This study aimed to investigate the role of CD200 in Treg-of-B (P) cell generation. To clarify the mechanisms, we used wild-type, STAT6 deficient, and IL-24 deficient T cells to generate Treg-of-B (P) cells, and antagonist antibodies (anti-CD200 and anti-IL-20RB), an agonist anti-CD200R antibody, CD39 inhibitors (ARL67156 and POM-1), a STAT6 inhibitor (AS1517499), and soluble IL-20RB were also applied. Our findings revealed that Peyer's patch B cells expressed CD200 to activate the CD200R on T cells and initiate the process of Treg-of-B (P) cells generation. CD200 and CD200R interaction triggers the phosphorylation of STAT6, which regulated the expression of CD200R, CD39, and IL-24 in T cells. CD39 regulated the expression of IL-24, which sustained the expression of CD223 and IL-10 and maintained the cell viability. In summary, the generation of Treg-of-B (P) cells by Peyer's patch B cells was through the CD200R-STAT6-CD39-IL-24 axis pathway.


Sujet(s)
Lymphocytes B , Facteur de transcription STAT-6 , Lymphocytes T régulateurs , Animaux , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Souris , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Facteur de transcription STAT-6/métabolisme , Souris de lignée C57BL , Récepteurs des orexines/métabolisme , Récepteurs des orexines/génétique , Antigènes CD/métabolisme , Antigènes CD/génétique , Antigènes CD/immunologie , Transduction du signal , Phosphorylation , Plaques de Peyer/immunologie , Plaques de Peyer/métabolisme , Plaques de Peyer/cytologie , Apyrase/métabolisme , Apyrase/immunologie , Glycoprotéines membranaires
18.
Stem Cell Rev Rep ; 20(5): 1357-1366, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38635127

RÉSUMÉ

Purinergic signaling is an ancient primordial signaling system regulating tissue development and specification of various types of stem cells. Thus, functional purinergic receptors are present in several types of cells in the body, including multiple populations of stem cells. However, one stem cell type that has not been evaluated for expression of purinergic receptors is very small embryonic stem cells (VSELs) isolated from postnatal tissues. Herein, we report that human umbilical cord blood (UCB) and murine bone marrow (BM) purified VSELs express mRNA for P1 and P2 purinergic receptors and CD39 and CD73 ectonucleotidases converting extracellular ATP (eATP) into its signaling metabolite extracellular adenosine (eAdo), that antagonizes eATP effects. More importantly, we demonstrate that human and murine VSELs respond by chemotaxis to eATP, and eAdo inhibits this migration. These responses to eATP are mediated by activation of Nlrp3 inflammasome, and exposure of VSELs to its specific inhibitor MCC950 abolished the chemotactic response to ATP. We conclude that purinergic signaling plays an essential, underappreciated role in the biology of these cells and their potential role in response to tissue/organ injuries.


Sujet(s)
Adénosine triphosphate , Apyrase , Mouvement cellulaire , Cellules souches embryonnaires , Humains , Adénosine triphosphate/métabolisme , Animaux , Souris , Cellules souches embryonnaires/métabolisme , Cellules souches embryonnaires/cytologie , Apyrase/métabolisme , Récepteurs purinergiques/métabolisme , 5'-Nucleotidase/métabolisme , 5'-Nucleotidase/génétique , Chimiotaxie , Antigènes CD/métabolisme , Antigènes CD/génétique , Sang foetal/cytologie , Sang foetal/métabolisme , Adénosine/métabolisme , Transduction du signal
19.
PLoS Pathog ; 20(4): e1012191, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38683845

RÉSUMÉ

An imbalance between suppressor and effector immune responses may preclude cure in chronic parasitic diseases. In the case of Trypanosoma cruzi infection, specialized regulatory Foxp3+ T (Treg) cells suppress protective type-1 effector responses. Herein, we investigated the kinetics and underlying mechanisms behind the regulation of protective parasite-specific CD8+ T cell immunity during acute T. cruzi infection. Using the DEREG mouse model, we found that Treg cells play a role during the initial stages after T. cruzi infection, restraining the magnitude of CD8+ T cell responses and parasite control. Early Treg cell depletion increased the frequencies of polyfunctional short-lived, effector T cell subsets, without affecting memory precursor cell formation or the expression of activation, exhaustion and functional markers. In addition, Treg cell depletion during early infection minimally affected the antigen-presenting cell response but it boosted CD4+ T cell responses before the development of anti-parasite effector CD8+ T cell immunity. Crucially, the absence of CD39 expression on Treg cells significantly bolstered effector parasite-specific CD8+ T cell responses, preventing increased parasite replication in T. cruzi infected mice adoptively transferred with Treg cells. Our work underscores the crucial role of Treg cells in regulating protective anti-parasite immunity and provides evidence that CD39 expression by Treg cells represents a key immunomodulatory mechanism in this infection model.


Sujet(s)
Antigènes CD , Apyrase , Lymphocytes T CD8+ , Maladie de Chagas , Lymphocytes T régulateurs , Trypanosoma cruzi , Animaux , Maladie de Chagas/immunologie , Lymphocytes T régulateurs/immunologie , Lymphocytes T CD8+/immunologie , Souris , Trypanosoma cruzi/immunologie , Antigènes CD/immunologie , Antigènes CD/métabolisme , Apyrase/immunologie , Apyrase/métabolisme , Souris de lignée C57BL , Modèles animaux de maladie humaine
20.
Nat Immunol ; 25(5): 834-846, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38561495

RÉSUMÉ

Cancer remains one of the leading causes of mortality worldwide, leading to increased interest in utilizing immunotherapy strategies for better cancer treatments. In the past decade, CD103+ T cells have been associated with better clinical prognosis in patients with cancer. However, the specific immune mechanisms contributing toward CD103-mediated protective immunity remain unclear. Here, we show an unexpected and transient CD61 expression, which is paired with CD103 at the synaptic microclusters of T cells. CD61 colocalization with the T cell antigen receptor further modulates downstream T cell antigen receptor signaling, improving antitumor cytotoxicity and promoting physiological control of tumor growth. Clinically, the presence of CD61+ tumor-infiltrating T lymphocytes is associated with improved clinical outcomes, mediated through enhanced effector functions and phenotype with limited evidence of cellular exhaustion. In conclusion, this study identified an unconventional and transient CD61 expression and pairing with CD103 on human immune cells, which potentiates a new target for immune-based cellular therapies.


Sujet(s)
Antigènes CD , Apyrase , Intégrines alpha , Récepteurs aux antigènes des cellules T , Transduction du signal , Animaux , Humains , Souris , Antigènes CD/métabolisme , Antigènes CD/immunologie , Lignée cellulaire tumorale , Cytotoxicité immunologique , Intégrines alpha/métabolisme , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Tumeurs/immunologie , Tumeurs/thérapie , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs aux antigènes des cellules T/immunologie , Transduction du signal/immunologie , Lymphocytes T cytotoxiques/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE