Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.776
Filtrer
1.
J Cancer Res Clin Oncol ; 150(6): 286, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38833021

RÉSUMÉ

BACKGROUND: Granzyme K (GZMK) is a crucial mediator released by immune cells to eliminate tumor cells, playing significant roles in inflammation and tumorigenesis. Despite its importance, the specific role of GZMK in breast cancer and its mechanisms are not well understood. METHODS: We utilized data from the TCGA and GEO databases and employed a range of analytical methods including GO, KEGG, GSEA, ssGSEA, and PPI to investigate the impact of GZMK on breast cancer. In vitro studies, including RT-qPCR, CCK-8 assay, cell cycle experiments, apoptosis assays, Celigo scratch assays, Transwell assays, and immunohistochemical methods, were conducted to validate the effects of GZMK on breast cancer cells. Additionally, Cox regression analysis integrating TCGA and our clinical data was used to develop an overall survival (OS) prediction model. RESULTS: Analysis of clinical pathological features revealed significant correlations between GZMK expression and lymph node staging, differentiation grade, and molecular breast cancer subtypes. High GZMK expression was associated with improved OS, progression-free survival (PFS), and recurrence-free survival (RFS), as confirmed by multifactorial Cox regression analysis. Functional and pathway enrichment analyses of genes positively correlated with GZMK highlighted involvement in lymphocyte differentiation, T cell differentiation, and T cell receptor signaling pathways. A robust association between GZMK expression and T cell presence was noted in the breast cancer tumor microenvironment (TME), with strong correlations with ESTIMATEScore (Cor = 0.743, P < 0.001), ImmuneScore (Cor = 0.802, P < 0.001), and StromalScore (Cor = 0.516, P < 0.001). GZMK also showed significant correlations with immune checkpoint molecules, including CTLA4 (Cor = 0.856, P < 0.001), PD-1 (Cor = 0.82, P < 0.001), PD-L1 (Cor = 0.56, P < 0.001), CD48 (Cor = 0.75, P < 0.001), and CCR7 (Cor = 0.856, P < 0.001). Studies indicated that high GZMK expression enhances patient responsiveness to immunotherapy, with higher levels observed in responsive patients compared to non-responsive ones. In vitro experiments confirmed that GZMK promotes cell proliferation, cell division, apoptosis, cell migration, and invasiveness (P < 0.05). CONCLUSION: Our study provides insights into the differential expression of GZMK in breast cancer and its potential mechanisms in breast cancer pathogenesis. Elevated GZMK expression is associated with improved OS and RFS, suggesting its potential as a prognostic marker for breast cancer survival and as a predictor of the efficacy of immunotherapy.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs du sein , Granzymes , Immunothérapie , Humains , Tumeurs du sein/anatomopathologie , Tumeurs du sein/immunologie , Tumeurs du sein/génétique , Tumeurs du sein/thérapie , Tumeurs du sein/mortalité , Femelle , Pronostic , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Immunothérapie/méthodes , Granzymes/métabolisme , Granzymes/génétique , Résultat thérapeutique , Adulte d'âge moyen , Microenvironnement tumoral/immunologie
2.
Front Immunol ; 15: 1392535, 2024.
Article de Anglais | MEDLINE | ID: mdl-38846935

RÉSUMÉ

The pivotal role of Granzyme B (GzmB) in immune responses, initially tied to cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells, has extended across diverse cell types and disease models. A number of studies have challenged conventional notions, revealing GzmB activity beyond apoptosis, impacting autoimmune diseases, inflammatory disorders, cancer, and neurotoxicity. Notably, the diverse functions of GzmB unfold through Perforin-dependent and Perforin-independent mechanisms, offering clinical implications and therapeutic insights. This review underscores the multifaceted roles of GzmB, spanning immunological and pathological contexts, which call for further investigations to pave the way for innovative targeted therapies.


Sujet(s)
Granzymes , Cellules tueuses naturelles , Perforine , Lymphocytes T cytotoxiques , Granzymes/métabolisme , Humains , Perforine/métabolisme , Animaux , Lymphocytes T cytotoxiques/immunologie , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Maladies auto-immunes/immunologie , Maladies auto-immunes/thérapie , Tumeurs/immunologie , Tumeurs/thérapie
3.
Gut Microbes ; 16(1): 2363020, 2024.
Article de Anglais | MEDLINE | ID: mdl-38841892

RÉSUMÉ

CD4+ T cells play a critical role in regulating autoimmune diseases, and intestinal microbial metabolites control various immune responses. Granzyme B (GzmB)-producing CD4+ T cells have been recently reported to participate in the pathogenesis of autoimmune diseases. Here, we found that GzmbB-deficient CD4+ T cells induced more severe colitis in Rag1-/- mice than wild-type (WT) CD4+ T cells. Germ-free (GF) mice exhibited a lower expression of GzmB in intestinal CD4+ T cells compared to specific pathogen-free (SPF) mice. Intestinal microbial metabolite butyrate increased GzmB expression in CD4+ T cells, especially in IL-10-producing Th1 cells, through HDAC inhibition and GPR43, but not GPR41 and GPR109a. Butyrate-treated GzmB-deficient CD4+ T cells demonstrated more severe colitis compared to butyrate-treated WT CD4+ T cells in the T cell transfer model. Butyrate altered intestinal microbiota composition, but altered microbiota did not mediate butyrate induction of intestinal CD4+ T cell expression of GzmB in mice. Blimp1 was involved in the butyrate induction of GzmB in IL-10-producing Th1 cells. Glucose metabolism, including glycolysis and pyruvate oxidation, mediated butyrate induction of GzmB in Th1 cells. In addition, we found that IKZF3 and NR2F6 regulated GzmB expression induced by butyrate. Together, our studies underscored the critical role of GzmB in mediating gut bacterial metabolite butyrate regulation of T cell tolerance at the mucosal surface.


Sujet(s)
Butyrates , Colite , Microbiome gastro-intestinal , Granzymes , Interleukine-10 , Souris de lignée C57BL , Lymphocytes auxiliaires Th1 , Animaux , Interleukine-10/métabolisme , Interleukine-10/génétique , Interleukine-10/immunologie , Lymphocytes auxiliaires Th1/immunologie , Souris , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Butyrates/métabolisme , Butyrates/pharmacologie , Granzymes/métabolisme , Colite/immunologie , Colite/microbiologie , Colite/métabolisme , Souris knockout , Facteur-1 liant le domaine de régulation positive I/métabolisme , Facteur-1 liant le domaine de régulation positive I/génétique , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Tolérance immunitaire , Protéines à homéodomaine
4.
Sci Rep ; 14(1): 13074, 2024 06 06.
Article de Anglais | MEDLINE | ID: mdl-38844784

RÉSUMÉ

While adaptive immune responses have been studied extensively in SLE (systemic lupus erythematosus), there is limited and contradictory evidence regarding the contribution of natural killer (NK) cells to disease pathogenesis. There is even less evidence about the role of NK cells in the more severe phenotype with juvenile-onset (J)SLE. In this study, analysis of the phenotype and function of NK cells in a large cohort of JSLE patients demonstrated that total NK cells, as well as perforin and granzyme A expressing NK cell populations, were significantly diminished in JSLE patients compared to age- and sex-matched healthy controls. The reduction in NK cell frequency was associated with increased disease activity, and transcriptomic analysis of NK populations from active and low disease activity JSLE patients versus healthy controls confirmed that disease activity was the main driver of differential NK cell gene expression. Pathway analysis of differentially expressed genes revealed an upregulation of interferon-α responses and a downregulation of exocytosis in active disease compared to healthy controls. Further gene set enrichment analysis also demonstrated an overrepresentation of the apoptosis pathway in active disease. This points to increased propensity for apoptosis as a potential factor contributing to NK cell deficiency in JSLE.


Sujet(s)
Cellules tueuses naturelles , Lupus érythémateux disséminé , Humains , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Lupus érythémateux disséminé/génétique , Lupus érythémateux disséminé/immunologie , Lupus érythémateux disséminé/anatomopathologie , Femelle , Mâle , Adolescent , Enfant , Phénotype , Granzymes/métabolisme , Granzymes/génétique , Perforine/métabolisme , Perforine/génétique , Apoptose/génétique , Transcriptome , Analyse de profil d'expression de gènes , Études cas-témoins
5.
Sci Rep ; 14(1): 12188, 2024 05 28.
Article de Anglais | MEDLINE | ID: mdl-38806640

RÉSUMÉ

Natural killer (NK) cells are closely associated with malignant tumor progression and metastasis. However, studies on their relevance in colorectal cancer (CRC) are limited. We aimed to comprehensively analyze the absolute counts, phenotypes, and function of circulating NK cells in patients with CRC using multiparametric flow cytometry. The distribution of NK cell subsets in the peripheral circulation of patients with CRC was significantly altered relative to the control group. This is shown by the decreased frequency and absolute count of CD56dimCD16+ NK cells with antitumor effects, contrary to the increased frequency of CD56bright NK and CD56dimCD16- NK cells with poor or ineffective antitumor effects. NK cells in patients with CRC were functionally impaired, with decreased intracellular interferon (IFN)-γ secretion and a significantly lower percentage of cell surface granzyme B and perforin expression. In addition, IFN-γ expression decreased significantly with the tumor stage progression. Based on a comprehensive analysis of the absolute counts, phenotypes, and functional markers of NK cells, we found an altered subset distribution and impaired function of circulating NK cells in patients with CRC.


Sujet(s)
Tumeurs colorectales , Granzymes , Interféron gamma , Cellules tueuses naturelles , Humains , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Tumeurs colorectales/immunologie , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Tumeurs colorectales/sang , Mâle , Femelle , Adulte d'âge moyen , Interféron gamma/métabolisme , Sujet âgé , Granzymes/métabolisme , Perforine/métabolisme , Antigènes CD56/métabolisme , Cytométrie en flux , Adulte
6.
BMC Cancer ; 24(1): 627, 2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38783281

RÉSUMÉ

BACKGROUND: CCR8-expressing regulatory T cells (Tregs) are selectively localized within tumors and have gained attention as potent suppressors of anti-tumor immunity. This study focused on CCR8+ Tregs and their interaction with CD8+ T cells in the tumor microenvironment of human lung cancer. We evaluated their spatial distribution impact on CD8+ T cell effector function, specifically granzyme B (GzmB) expression, and clinical outcomes. METHODS: A total of 81 patients with lung squamous cell carcinoma (LSCC) who underwent radical surgical resection without preoperative treatment were enrolled. Histological analyses were performed, utilizing an automated image analysis system for double-stained immunohistochemistry assays of CCR8/Foxp3 and GzmB/CD8. We investigated the association of CCR8+ Tregs and GzmB+ CD8+ T cells in tumor tissues and further evaluated the prognostic impact of their distribution profiles. RESULTS: Histological evaluation using the region of interest (ROI) protocol showed that GzmB expression levels in CD8+ T cells were decreased in areas with high infiltration of CCR8+ Tregs, suggesting a suppressive effect of CCR8+ Tregs on T cell cytotoxicity in the local tumor microenvironment. Analysis of the association with clinical outcomes showed that patients with more CCR8+ Tregs and lower GzmB expression, represented by a low GzmB/CCR8 ratio, had worse progression-free survival. CONCLUSIONS: Our data suggest that local CCR8+ Treg accumulation is associated with reduced CD8+ T cell cytotoxic activity and poor prognosis in LSCC patients, highlighting the biological role and clinical significance of CCR8+ Tregs in the tumor microenvironment. The GzmB/CCR8 ratio may be a useful prognostic factor for future clinical applications in LSCC.


Sujet(s)
Lymphocytes T CD8+ , Granzymes , Tumeurs du poumon , Récepteurs CCR8 , Lymphocytes T régulateurs , Microenvironnement tumoral , Humains , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/mortalité , Tumeurs du poumon/métabolisme , Tumeurs du poumon/chirurgie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Pronostic , Femelle , Mâle , Récepteurs CCR8/métabolisme , Récepteurs CCR8/immunologie , Granzymes/métabolisme , Microenvironnement tumoral/immunologie , Sujet âgé , Adulte d'âge moyen , Carcinome épidermoïde/immunologie , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/métabolisme , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Sujet âgé de 80 ans ou plus , Adulte
7.
Nat Commun ; 15(1): 3937, 2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38729924

RÉSUMÉ

Human natural killer (NK) cell-based therapies are under assessment for treating various cancers, but cryopreservation reduces both the recovery and function of NK cells, thereby limiting their therapeutic feasibility. Using cryopreservation protocols optimized for T cells, here we find that ~75% of NK cells die within 24 h post-thaw, with the remaining cells displaying reduced cytotoxicity. Using CRISPR-Cas9 gene editing and confocal microscopy, we find that cryopreserved NK cells largely die via apoptosis initiated by leakage of granzyme B from cytotoxic vesicles. Pretreatment of NK cells with a combination of Interleukins-15 (IL-15) and IL-18 prior to cryopreservation improves NK cell recovery to ~90-100% and enables equal tumour control in a xenograft model of disseminated Raji cell lymphoma compared to non-cryopreserved NK cells. The mechanism of IL-15 and IL-18-induced protection incorporates two mechanisms: a transient reduction in intracellular granzyme B levels via degranulation, and the induction of antiapoptotic genes.


Sujet(s)
Apoptose , Cryoconservation , Granzymes , Interleukine-15 , Interleukine-18 , Cellules tueuses naturelles , Granzymes/métabolisme , Interleukine-15/métabolisme , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Humains , Interleukine-18/métabolisme , Animaux , Cryoconservation/méthodes , Souris , Lignée cellulaire tumorale , Systèmes CRISPR-Cas
8.
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
9.
Yakugaku Zasshi ; 144(5): 475-481, 2024.
Article de Japonais | MEDLINE | ID: mdl-38692920

RÉSUMÉ

Zinc is one of the essential trace elements, and is involved in various functions in the body. Zinc deficiency is known to cause immune abnormalities, but the mechanism is not fully understood. Therefore, we focused our research on tumor immunity to elucidate the effect of zinc on colorectal cancer and its mechanisms. Mice were treated with azoxymethane (AOM) and dextran sodium sulfate (DSS) to develop colorectal cancer, then the relationship between zinc content in the diet and the number and area of tumors in the colon was observed. The number of tumors in the colon was significantly higher in the no-zinc-added diet group compared to the normal zinc intake group, and about half the number in the high-zinc-intake group compared to the normal-zinc-intake group. In T-cell-deficient mice, the number of tumors in the high-zinc-intake group was similar to that in the normal-zinc-intake group, suggesting that the inhibitory effect of zinc was dependent on T cells. Furthermore, we found that the amount of granzyme B transcript released by cytotoxic T cells upon antigen stimulation was significantly increased by the addition of zinc. We also showed that granzyme B transcriptional activation by zinc addition was dependent on calcineurin activity. Collectively, we have shown that zinc exerts its tumor-suppressive effect by acting on cytotoxic T cells, the center of cellular immunity, and that it increases the transcription of granzyme B, one of the key molecules involved in tumor immunity. In this symposium, we would like to introduce our latest data on the relationship between zinc and tumor immunity.


Sujet(s)
Tumeurs colorectales , Immunité cellulaire , Zinc , Animaux , Humains , Souris , Oxyde de diméthyl-diazène , Tumeurs colorectales/immunologie , Tumeurs colorectales/étiologie , Tumeurs colorectales/prévention et contrôle , Modèles animaux de maladie humaine , Granzymes/métabolisme , Lymphocytes T cytotoxiques/immunologie
10.
ACS Infect Dis ; 10(6): 2108-2117, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38819300

RÉSUMÉ

Understanding how the host immune system engages complex pathogens is essential to developing therapeutic strategies to overcome their virulence. While granzymes are well understood to trigger apoptosis in infected host cells or bacteria, less is known about how the immune system mobilizes individual granzyme species in vivo to combat diverse pathogens. Toward the goal of studying individual granzyme function directly in vivo, we previously developed a new class of radiopharmaceuticals termed "restricted interaction peptides (RIPs)" that detect biochemically active endoproteases using positron emission tomography (PET). In this study, we showed that secreted granzyme B proteolysis in response to diverse viral and bacterial pathogens could be imaged with [64Cu]Cu-GRIP B, a RIP that specifically targets granzyme B. Wild-type or germline granzyme B knockout mice were instilled intranasally with the A/PR/8/34 H1N1 influenza A strain to generate pneumonia, and granzyme B production within the lungs was measured using [64Cu]Cu-GRIP B PET/CT. Murine myositis models of acute bacterial (E. coli, P. aeruginosa, K. pneumoniae, and L. monocytogenes) infection were also developed and imaged using [64Cu]Cu-GRIP B. In all cases, the mice were studied in vivo using mPET/CT and ex vivo via tissue-harvesting, gamma counting, and immunohistochemistry. [64Cu]Cu-GRIP B uptake was significantly higher in the lungs of wild-type mice that received A/PR/8/34 H1N1 influenza A strain compared to mice that received sham or granzyme B knockout mice that received either treatment. In wild-type mice, [64Cu]Cu-GRIP B uptake was significantly higher in the infected triceps muscle versus normal muscle and the contralateral triceps inoculated with heat killed bacteria. In granzyme B knockout mice, [64Cu]Cu-GRIP B uptake above the background was not observed in the infected triceps muscle. Interestingly, live L. monocytogenes did not induce detectable granzyme B on PET, despite prior in vitro data, suggesting a role for granzyme B in suppressing their pathogenicity. In summary, these data show that the granzyme response elicited by diverse human pathogens can be imaged using PET. These results and data generated via additional RIPs specific for other granzyme proteases will allow for a deeper mechanistic study analysis of their complex in vivo biology.


Sujet(s)
Granzymes , Souris knockout , Animaux , Granzymes/métabolisme , Souris , Tomographie par émission de positons/méthodes , Tomographie par émission de positons couplée à la tomodensitométrie , Radio-isotopes du cuivre , Femelle , Souris de lignée C57BL , Infections bactériennes/imagerie diagnostique , Infections bactériennes/immunologie , Modèles animaux de maladie humaine , Poumon/imagerie diagnostique , Poumon/microbiologie , Poumon/immunologie , Radiopharmaceutiques , Infections à Orthomyxoviridae/immunologie
11.
Front Immunol ; 15: 1371743, 2024.
Article de Anglais | MEDLINE | ID: mdl-38646541

RÉSUMÉ

Granzymes are a family of serine proteases, composed of five human members: GA, B, H, M and K. They were first discovered in the 1980s within cytotoxic granules released during NK cell- and T cell-mediated killing. Through their various proteolytic activities, granzymes can trigger different pathways within cells, all of which ultimately lead to the same result, cell death. Over the years, the initial consideration of granzymes as mere cytotoxic mediators has changed due to surprising findings demonstrating their expression in cells other than immune effectors as well as new intracellular and extracellular activities. Additional roles have been identified in the extracellular milieu, following granzyme escape from the immunological synapse or their release by specific cell types. Outside the cell, granzyme activities mediate extracellular matrix alteration via the degradation of matrix proteins or surface receptors. In certain contexts, these processes are essential for tissue homeostasis; in others, excessive matrix degradation and extensive cell death contribute to the onset of chronic diseases, inflammation, and autoimmunity. Here, we provide an overview of both the physiological and pathological roles of granzymes, highlighting their utility while also recognizing how their unregulated presence can trigger the development and/or worsening of diseases.


Sujet(s)
Granzymes , Humains , Granzymes/métabolisme , Animaux , Matrice extracellulaire/métabolisme , Matrice extracellulaire/immunologie , Inflammation/immunologie , Cellules tueuses naturelles/immunologie
12.
PLoS One ; 19(4): e0302406, 2024.
Article de Anglais | MEDLINE | ID: mdl-38635551

RÉSUMÉ

Natural killer (NK) cells play a crucial role in immunosurveillance independent of antigen presentation, which is regulated by signal balance via activating and inhibitory receptors. The anti-tumor activity of NK cells is largely dependent on signaling from target recognition to cytolytic degranulation; however, the underlying mechanism remains unclear, and NK cell cytotoxicity is readily impaired by tumor cells. Understanding the activation mechanism is necessary to overcome the immune evasion mechanism, which remains an obstacle in immunotherapy. Because calcium ions are important activators of NK cells, we hypothesized that electrical stimulation could induce changes in intracellular Ca2+ levels, thereby improving the functional potential of NK cells. In this study, we designed an electrical stimulation system and observed a correlation between elevated Ca2+ flux induced by electrical stimulation and NK cell activation. Breast cancer MCF-7 cells co-cultured with electrically stimulated KHYG-1 cells showed a 1.27-fold (0.5 V/cm) and 1.55-fold (1.0 V/cm) higher cytotoxicity, respectively. Electrically stimulated KHYG-1 cells exhibited a minor increase in Ca2+ level (1.31-fold (0.5 V/cm) and 1.11-fold (1.0 V/cm) higher), which also led to increased gene expression of granzyme B (GZMB) by 1.36-fold (0.5 V/cm) and 1.58-fold (1.0 V/cm) by activating Ca2+-dependent nuclear factor of activated T cell 1 (NFAT1). In addition, chelating Ca2+ influx with 5 µM BAPTA-AM suppressed the gene expression of Ca2+ signaling and lytic granule (granzyme B) proteins by neutralizing the effects of electrical stimulation. This study suggests a promising immunotherapeutic approach without genetic modifications and elucidates the correlation between cytolytic effector function and intracellular Ca2+ levels in electrically stimulated NK cells.


Sujet(s)
Calcium , Tumeurs , Humains , Granzymes/métabolisme , Calcium/métabolisme , Cellules tueuses naturelles , Activation des lymphocytes , Tumeurs/métabolisme , Cytotoxicité immunologique
13.
Front Immunol ; 15: 1349749, 2024.
Article de Anglais | MEDLINE | ID: mdl-38629077

RÉSUMÉ

Background: Respiratory syncytial virus (RSV) is the most common cause of viral lower respiratory tract infections (LRTIs) in young children around the world and an important cause of LRTI in the elderly. The available treatments and FDA-approved vaccines for RSV only lessen the severity of the infection and are recommended for infants and elderly people. Methods: We focused on developing a broad-spectrum vaccine that activates the immune system to directly combat RSV. The objective of this study is to identify CD4+ and CD8+ T-cell epitopes using an immunoinformatics approach to develop RSV vaccines. The efficacy of these peptides was validated through in-vitro and in-vivo studies involving healthy and diseased animal models. Results: For each major histocompatibility complex (MHC) class-I and II, we found three epitopes of RSV proteins including F, G, and SH with an antigenic score of >0.5 and a projected SVM score of <5. Experimental validation of these peptides on female BALB/c mice was conducted before and after infection with the RSV A2 line 19f. We found that the 3RVMHCI (CD8+) epitope of the F protein showed significant results of white blood cells (19.72 × 103 cells/µl), neutrophils (6.01 × 103 cells/µl), lymphocytes (12.98 × 103 cells/µl), IgG antibodies (36.9 µg/ml), IFN-γ (86.96 ng/L), and granzyme B (691.35 pg/ml) compared to control at the second booster dose of 10 µg. Similarly, 4RVMHCII (CD4+) of the F protein substantially induced white blood cells (27.08 × 103 cells/µl), neutrophils (6.58 × 103 cells/µl), lymphocytes (16.64 × 103 cells/µl), IgG antibodies (46.13 µg/ml), IFN-γ (96.45 ng/L), and granzyme B (675.09 pg/ml). In-vitro studies showed that 4RVMHCII produced a significant level of antibodies in sera on day 45 comparable to mice infected with the virus. 4RVMHCII also induced high IFN-γ and IL-2 secretions on the fourth day of the challenge compared to the preinfectional stage. Conclusion: In conclusion, epitopes of the F protein showed considerable immune response and are suitable for further validation.


Sujet(s)
Déterminants antigéniques des lymphocytes T , Infections à virus respiratoire syncytial , Virus respiratoire syncytial humain , Sujet âgé , Animaux , Enfant , Enfant d'âge préscolaire , Femelle , Humains , Nourrisson , Souris , Anticorps antiviraux , Lymphocytes T CD4+ , Lymphocytes T CD8+ , Déterminants antigéniques des lymphocytes T/métabolisme , Granzymes , Immunoglobuline G , Peptides , Infections à virus respiratoire syncytial/immunologie , Virus respiratoire syncytial humain/métabolisme
14.
Immun Inflamm Dis ; 12(4): e1255, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38652012

RÉSUMÉ

BACKGROUND: Natural killer (NK) cells, CD3- lymphocytes, are critical players in cancer immune surveillance. This study aimed to assess two types of CD3- NK cell classifications (subsets), that is, convectional subsets (based on CD56 and CD16 expression) and new subsets (based on CD56, CD27, and CD11b expression), and their functional molecules in the peripheral blood of patients with breast cancer (BC) in comparison with healthy donors (HDs). METHODS: Thirty untreated females with BC and 20 age-matched healthy women were enrolled. Peripheral blood samples were collected and directly incubated with fluorochrome-conjugated antibodies against CD3, CD56, CD16, CD27, CD11b, CD96, NKG2C, NKG2D, NKp44, CXCR3, perforin, and granzyme B. Red blood cells were then lysed using lysing solution, and the stained cells were acquired on four-color flow cytometer. RESULT: Our results indicated 15% of lymphocytes in peripheral blood of patients with BC and HDs had NK cells phenotype. However, the frequency of total NK cells (CD3-CD56+), and NK subsets (based on conventional and new classifications) was not significantly different between patients and HDs. We observed mean fluorescent intensity (MFI) of CXCR3 in total NK cells (p = .02) and the conventional cytotoxic (CD3-CD56dim CD16+) NK cells (p = .03) were significantly elevated in the patients with BC compared to HDs. Despite this, the MFI of granzyme B expression in conventional regulatory (CD3-CD56brightCD16- /+) NK cells and CD3-CD56-CD16+ NK cells (p = .03 and p = .004, respectively) in the patients was lower than healthy subjects. CONCLUSION: The higher expression of chemokine receptor CXCR3 on total NK cells in patients with BC may be associated with increased chemotaxis-related NK cell infiltration. However, lower expression of granzyme B in conventional regulatory NK cells and CD3-CD56-CD16+ NK cells in the patients compared to HDs suggests reduced cytotoxic activity of the NK cells in BC. These results might demonstrate accumulating NK subsets with a dysfunctional phenotype in the peripheral blood of patients with BC.


Sujet(s)
Tumeurs du sein , Cellules tueuses naturelles , Humains , Femelle , Tumeurs du sein/immunologie , Tumeurs du sein/sang , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Adulte d'âge moyen , Adulte , Sujet âgé , Cytométrie en flux , Immunophénotypage , Sous-populations de lymphocytes/immunologie , Sous-populations de lymphocytes/métabolisme , Granzymes/sang , Antigènes CD/sang , Antigènes CD/immunologie
15.
Toxicon ; 243: 107723, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38663519

RÉSUMÉ

Tumor-associated macrophages within the tumor microenvironment play an immunosuppressive role by promoting tumor growth and immune evasion. Macrophages are highly plastic and can be stimulated to adopt an anti-tumor M1 phenotype. In this study, we used microcystin-LR (MC-LR), a cyclic heptapeptide produced by cyanobacteria, to induce in vitro macrophage innate immunity and transition into the anti-tumor M1 phenotype. MC-LR was also tested in vivo in a mouse model of colorectal cancer. An intraperitoneal injection of MC-LR increased the proportion of CD86⁺ M1 macrophages and triggered the maturation of CD11c⁺ dendritic cells within tumor tissues. MC-LR combined with the chemotherapeutic drug oxaliplatin significantly inhibited tumor growth in vivo. Flow cytometry analysis revealed increased infiltration of activated cytotoxic (CD8⁺, PD-1⁺) T-cells and anti-tumor cytokines (IFNγ and Granzyme B) in the tumor tissues of the combination therapy group, suggesting that this may be the primary mechanism behind the anti-tumor effect of the combination treatment. These findings indicate that MC-LR regulates the immune stimulation of macrophage polarization and dendritic cell maturation, effectively reversing tumor immunosuppression, activating an anti-tumor immune response, and enhancing tumor therapy.


Sujet(s)
Antinéoplasiques , Macrophages , Oxaliplatine , Oxaliplatine/pharmacologie , Oxaliplatine/usage thérapeutique , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Macrophages/immunologie , Macrophages/physiologie , Synergie des médicaments , Animaux , Souris , Lignée cellulaire tumorale , Tumeurs du rectum/traitement médicamenteux , Cellules dendritiques , Granzymes/métabolisme , Interféron gamma/métabolisme , Immunité innée , Cyanobactéries
16.
Nat Rev Rheumatol ; 20(6): 361-376, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38689140

RÉSUMÉ

Granzymes (granule-secreted enzymes) are a family of serine proteases that have been viewed as redundant cytotoxic enzymes since their discovery more than 30 years ago. Predominantly produced by cytotoxic lymphocytes and natural killer cells, granzymes are delivered into the cytoplasm of target cells through immunological synapses in cooperation with the pore-forming protein perforin. After internalization, granzymes can initiate cell death through the cleavage of intracellular substrates. However, evidence now also demonstrates the existence of non-cytotoxic, pro-inflammatory, intracellular and extracellular functions that are granzyme specific. Under pathological conditions, granzymes can be produced and secreted extracellularly by immune cells as well as by non-immune cells. Depending on the granzyme, accumulation in the extracellular milieu might contribute to inflammation, tissue injury, impaired wound healing, barrier dysfunction, osteoclastogenesis and/or autoantigen generation.


Sujet(s)
Granzymes , Inflammation , Rhumatismes , Granzymes/métabolisme , Humains , Inflammation/immunologie , Rhumatismes/immunologie , Rhumatismes/enzymologie , Animaux
17.
J Control Release ; 369: 658-667, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38604384

RÉSUMÉ

Granzyme B (GrB)-based immunotherapy is of interest for cancer treatment. However, insufficient cellular uptake and a lack of targeting remain challenges to make use of GrB for solid tumour therapy. As GrB induced cell death requires the help of perforin (PFN), we designed a system (nGPM) for the co-delivery of GrB and PFN. Therefore, GrB and PFN were loaded in a porous polymeric nanocapsule rich in acetylcholine analogues and matrix metalloproteinase-2 (MMP-2) responsive peptides. The neutrally charged nGPM nanocapsules showed as long circulating time and accumulated at the tumour sites. Once in the tumour the outside shell of nanocapsules became degraded by overexpressed MMP-2 proteases, resulting in the release of GrB and PFN. We found that the PFN complex formed small pores on the surface of tumour cells which allow GrB to enter the cytoplasm of tumour cells inducing cell apoptosis and tumour suppression significantly.


Sujet(s)
Granzymes , Nanocapsules , Perforine , Granzymes/métabolisme , Nanocapsules/composition chimique , Animaux , Perforine/métabolisme , Humains , Lignée cellulaire tumorale , Tumeurs/traitement médicamenteux , Tumeurs/thérapie , Tumeurs/immunologie , Apoptose/effets des médicaments et des substances chimiques , Matrix metalloproteinase 2/métabolisme , Immunothérapie/méthodes , Souris de lignée BALB C , Femelle , Souris
18.
J Immunol ; 212(11): 1722-1732, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38607279

RÉSUMÉ

An imbalance between proinflammatory and regulatory processes underlies autoimmune disease pathogenesis. We have shown that acute relapses of multiple sclerosis are characterized by a deficit in the immune suppressive ability of CD8+ T cells. These cells play an important immune regulatory role, mediated in part through cytotoxicity (perforin [PRF]/granzyme [GZM]) and IFNγ secretion. In this study, we further investigated the importance of IFNγ-, GZMB-, PRF1-, and LYST-associated pathways in CD8+ T cell-mediated suppression. Using the CRISPR-Cas9 ribonucleoprotein transfection system, we first optimized efficient gene knockout while maintaining high viability in primary bulk human CD8+ T cells. Knockout was confirmed through quantitative real-time PCR assays in all cases, combined with flow cytometry where appropriate, as well as confirmation of insertions and/or deletions at genomic target sites. We observed that the knockout of IFNγ, GZMB, PRF1, or LYST, but not the knockout of IL4 or IL5, resulted in significantly diminished in vitro suppressive ability in these cells. Collectively, these results reveal a pivotal role for these pathways in CD8+ T cell-mediated immune suppression and provide important insights into the biology of human CD8+ T cell-mediated suppression that could be targeted for immunotherapeutic intervention.


Sujet(s)
Lymphocytes T CD8+ , Granzymes , Interféron gamma , Perforine , Humains , Lymphocytes T CD8+/immunologie , Interféron gamma/immunologie , Interféron gamma/métabolisme , Perforine/génétique , Perforine/métabolisme , Granzymes/métabolisme , Granzymes/génétique , Systèmes CRISPR-Cas , Sclérose en plaques/immunologie , Sclérose en plaques/génétique , Techniques de knock-out de gènes , Cellules cultivées
19.
Methods Mol Biol ; 2782: 65-80, 2024.
Article de Anglais | MEDLINE | ID: mdl-38622392

RÉSUMÉ

Maintaining immune homeostasis is instrumental for host health. Immune cells, such as T cells, are instrumental for the eradication of pathogenic bacteria, fungi and viruses. Furthermore, T cells also play a major role in the fight against cancer. Through the formation of immunological memory, a pool of antigen-experienced T cells remains in the body to rapidly protect the host upon reinfection or retransformation. In order to perform their protective function, T cells produce cytolytic molecules, such as granzymes and perforin, and cytokines such as interferon γ and tumor necrosis factor α. Recently, it has become evident that posttranscriptional regulatory events dictate the kinetics and magnitude of cytokine production by murine and human CD8+ T cells. Here, the recent literature regarding the role posttranscriptional regulation plays in maintaining immune homeostasis of antigen-experienced CD8+ T cells is reviewed.


Sujet(s)
Lymphocytes T CD8+ , Glycoprotéines membranaires , Humains , Animaux , Souris , Perforines , Cytokines , Perforine , Granzymes , Homéostasie
20.
Methods Mol Biol ; 2782: 209-226, 2024.
Article de Anglais | MEDLINE | ID: mdl-38622405

RÉSUMÉ

T cells are instrumental in protecting the host against invading pathogens and the development of cancer. To do so, they produce effector molecules such as granzymes, interleukins, interferons, and perforin. For the development and immunomonitoring of therapeutic applications such as cell-based therapies and vaccines, assessing T cell effector function is paramount. This can be achieved through various methods, such as 51Cr release assays, flow cytometry, and enzyme-linked immune absorbent spot (ELISpot) assays. For T cell ELISpots, plates are coated with antibodies directed against the effector molecule of interest (e.g., IFN-g). Subsequently, peripheral blood mononuclear cells (PBMCs) or isolated T cells are cultured on the plate together with stimuli of choice, and the production of effector molecules is visualized via labeled detection antibodies. For clinical studies, ELISpot is currently the gold standard to determine antigen-specific T cell frequencies. In contrast to 51Cr release assays, ELISpot allows for the exact enumeration of responding T cells, and compared to flow cytometry, ELISpot is more cost-effective and high throughput. Here, we optimize and describe, in a step-by-step fashion, how to perform a controlled IFN-γ ELISpot experiment to determine the frequency of responding or antigen-specific T cells in healthy human volunteers. Of note, this protocol can also be employed to assess the frequency of antigen-specific T cells induced in, e.g., vaccination studies or present in cellular products.


Sujet(s)
Agranulocytes , Lymphocytes T , Humains , Test ELISpot/méthodes , Antigènes , Granzymes , Test ELISA/méthodes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...