Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.424
Filter
1.
Nat Commun ; 15(1): 8722, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39379371

ABSTRACT

Stevens-Johnson syndrome and toxic epidermal necrolysis (SJS/TEN) is a rare but life-threatening cutaneous drug reaction mediated by human leukocyte antigen (HLA) class I-restricted CD8+ T cells. For unbiased assessment of cellular immunopathogenesis, here we perform single-cell (sc) transcriptome, surface proteome, and T cell receptor (TCR) sequencing on unaffected skin, affected skin, and blister fluid from 15 SJS/TEN patients. From 109,888 cells, we identify 15 scRNA-defined subsets. Keratinocytes express markers indicating HLA class I-restricted antigen presentation and appear to trigger the proliferation of and killing by cytotoxic CD8+ tissue-resident T cells that express granulysin, granzyme B, perforin, LAG3, CD27, and LINC01871, and signal through the PKM, MIF, TGFß, and JAK-STAT pathways. In affected tissue, cytotoxic CD8+ T cells express private expanded and unexpanded TCRαß that are absent or unexpanded in unaffected skin, and mixed populations of macrophages and fibroblasts express pro-inflammatory markers or those favoring repair. This data identifies putative cytotoxic TCRs and therapeutic targets.


Subject(s)
CD8-Positive T-Lymphocytes , Keratinocytes , Receptors, Antigen, T-Cell , Single-Cell Analysis , Stevens-Johnson Syndrome , Humans , Stevens-Johnson Syndrome/immunology , Stevens-Johnson Syndrome/genetics , Single-Cell Analysis/methods , Keratinocytes/immunology , Keratinocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/genetics , Skin/immunology , Skin/pathology , T-Lymphocytes, Cytotoxic/immunology , Granzymes/metabolism , Granzymes/genetics , Transcriptome , Male , Perforin/metabolism , Perforin/genetics , Female , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/metabolism , Antigens, Differentiation, T-Lymphocyte/immunology , Macrophages/immunology , Macrophages/metabolism
2.
Gut Microbes ; 16(1): 2401649, 2024.
Article in English | MEDLINE | ID: mdl-39388633

ABSTRACT

The microbiota-associated factors that affect host susceptibility and adaptive immunity to influenza A virus (IAV) infection have not been fully elucidated. By comparing the microbiota composition between survivors and mice that succumbed to IAV strain PR8 infection, we identified that the commensal bacterium Blautia coccoides protects antibiotics (Abx)-treated or germ-free (GF) mice from PR8 infection by inducing functionally optimal virus-specific CD8+ T cell responses. Administration of exogenous acetate reproduced the protective effect of B. coccoides monocolonization in Abx and GF mice, enhancing oxidative phosphorylation and glycolysis as well as secretion of IFN-γ and granzyme B in virus-specific CD8+ T cells, dependent on GPR43 signaling and acetyl-CoA synthetase 2. Thus, we have demonstrated that microbiota-derived acetate possesses an antiviral effect that induces an optimal virus-specific CD8+ T cell response to IAV PR8 infection via GPR43-dependent metabolic reprogramming.


Subject(s)
Acetates , CD8-Positive T-Lymphocytes , Gastrointestinal Microbiome , Influenza A virus , Mice, Inbred C57BL , Orthomyxoviridae Infections , Receptors, G-Protein-Coupled , Animals , CD8-Positive T-Lymphocytes/immunology , Mice , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Acetates/metabolism , Acetates/pharmacology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/metabolism , Influenza A virus/immunology , Gastrointestinal Microbiome/drug effects , Granzymes/metabolism , Interferon-gamma/metabolism , Interferon-gamma/immunology , Anti-Bacterial Agents/pharmacology , Glycolysis/drug effects , Metabolic Reprogramming
3.
Nat Commun ; 15(1): 8791, 2024 Oct 10.
Article in English | MEDLINE | ID: mdl-39389969

ABSTRACT

To tackle the clinical challenge of noninvasively assessing immunotherapy efficacy in patients, here we used positron emission tomography (PET) with 68Ga-grazytracer, which targets granzyme B, a crucial effector molecule secreted by activated CD8+ T cells. In this phase 1/2 clinical trial (NCT05000372) involving a diverse cohort of 24 patients with solid tumors and lymphomas who received immunotherapies, including immune checkpoint inhibitors (either alone or with chemotherapies) and chimeric antigen receptor-T cell therapy, we examined the in vivo behaviors of 68Ga-grazytracer. Primary endpoints were safety, biodistribution, granzyme B specificity, and the predictive utility of 68Ga-grazytracer, while secondary endpoint was the relationship between 68Ga-grazytracer uptake and tumor immune phenotype. 68Ga-grazytracer exhibited a safe profile and specifically targeted granzyme B in patients. 68Ga-grazytracer PET showed superior predictive value for short-term prognosis and progression-free survival than those of conventional assessment criteria, including RECIST 1.1 and PERCIST. Moreover, the uptake of 68Ga-grazytracer in tumors was significantly higher in those with a "non-desert" immune phenotype than those with an immune "desert" phenotype, thereby meeting the primary and secondary endpoints of this trial. Collectively, we successfully visualized CD8+ T cell effector function in humans using 68Ga-grazytracer PET, offering insights for enhancing immunotherapy assessment, patient stratification and treatment planning.


Subject(s)
Granzymes , Immunotherapy , Lymphoma , Positron-Emission Tomography , Humans , Granzymes/metabolism , Positron-Emission Tomography/methods , Female , Lymphoma/diagnostic imaging , Lymphoma/therapy , Lymphoma/immunology , Male , Middle Aged , Aged , Immunotherapy/methods , Adult , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/diagnostic imaging , CD8-Positive T-Lymphocytes/immunology , Gallium Radioisotopes , Immune Checkpoint Inhibitors/therapeutic use , Progression-Free Survival , Tissue Distribution
4.
Anticancer Res ; 44(10): 4537-4542, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39348967

ABSTRACT

BACKGROUND/AIM: Granzyme B (GZMB) is mainly produced by natural killer (NK) cells and activated CD8-positive T cells to induce tumor cell apoptosis. We analyzed the significance of GZMB expression in gastric cancer (GC) tissues from patients with pathological (p)Stage II/III GC after curative resection. PATIENTS AND METHODS: Patients with pStage II/III GC who received curative resection (n=253) were included and the expression levels of GZMB in GC tissues and in the adjacent normal mucosa were measured using quantitative real-time polymerase chain reaction. The expression levels in GC tissues and clinicopathological features and overall survival (OS) were compared in these patients. RESULTS: GZMB expression levels were significantly higher in GC tissues than in the adjacent normal mucosa. GZMB expression levels in GC tissues were not associated with any clinicopathological features. The 5-year OS rate in the high-GZMB expression group was significantly better than that in the low-expression group (5-year survival rate 72.0% vs. 55.7%; p=0.009). Furthermore, on multivariate analysis, high-GZMB expression was an independent factor for better OS (hazard ratio=0.652; 95% confidence interval=0.432-0.987; p=0.043). CONCLUSION: In patients with locally advanced GC after curative resection, GZMB expression in GC tissue may be a useful prognostic marker.


Subject(s)
Gastrectomy , Granzymes , Neoplasm Staging , Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Granzymes/genetics , Granzymes/metabolism , Male , Female , Middle Aged , Aged , Prognosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Adult , Clinical Relevance
5.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(8): 1529-1536, 2024 Aug 20.
Article in Chinese | MEDLINE | ID: mdl-39276048

ABSTRACT

OBJECTIVE: To explore the correlation of baseline CCL19+ dendritic cell (CCL19+ DC) infiltration in lung adenocarcinoma microenvironment with immunotherapy efficacy and CD8+ T cell infiltration. METHODS: We retrospectively analyzed the data of patients with lung adenocarcinoma hospitalized at First Affiliated Hospital of Henan University of Science and Technology from January, 2020 to December, 2023, and collected tissue samples from 96 patients undergoing immunotherapy for assessing CCL19+ DC and CD8+ T cell infiltration using immunofluorescence assay. We evaluated the predictive value of baseline CCL19+ DCs for patient responses to immunotherapy using receiver-operating characteristics (ROC) curves and analyzed the correlations of baseline CCL19+ DC expression with immunotherapy efficacy and CD8+ T cell and cytotoxic T lymphocyte (CTL) infiltrations. In co-culture systems of lung adenocarcinoma PC9 cells, CD8+ T cells and DCs (overexpressing CCL19 with or without anti PD-1 antibody treatment), the expressions of granzyme B, perforin, IFN-γ, and Ki-67 in T cells were analyzed using flow cytometry. RESULTS: The patients with partial or complete remission following immunotherapy had a significantly higher baseline CCL19+ DC infiltration level in lung adenocarcinoma tissues than those with poor responses. CCL19+ DC infiltration had an area under ROC curve of 0.785, a sensitivity of 75.6%, and a specificity of 62.8% for predicting immunotherapy efficacy. The expression of CD8+ T cell surface molecules Granzyme B (P<0.01), Perforin (P<0.01), IFN-γ (P<0.01) and Ki-67 (P<0.001) in patients with high expression of CCL19+ DC were higher than those in patients with low expression of CCL19+ DC. The baseline CCL19+ DC infiltration level was positively correlated with immunotherapy efficacy (P=0.003), CTL infiltration of (r=0.6657, P<0.001) and CD8+ T cell infiltration (P=0.007). In the co-cultured cells, CCL19 overexpression combined with anti-PD1 treatment of the DCs more strongly enhanced cytotoxicity and proliferation of CD8+ T lymphocytes than either of the single treatments (P<0.01 or 0.001). CONCLUSION: The baseline CCL19+ DC infiltration level in lung adenocarcinoma microenvironment is positively correlated with immunotherapy efficacy and CTL infiltration and can thus predict the response to immunotherapy.


Subject(s)
Adenocarcinoma of Lung , Chemokine CCL19 , Dendritic Cells , Immunotherapy , Lung Neoplasms , Humans , Dendritic Cells/immunology , Lung Neoplasms/therapy , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Chemokine CCL19/metabolism , Immunotherapy/methods , Adenocarcinoma of Lung/therapy , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/pathology , Retrospective Studies , Tumor Microenvironment/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Male , Middle Aged , Granzymes/metabolism
6.
J Cell Mol Med ; 28(17): e18535, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39267250

ABSTRACT

Luteolin, a commonly used traditional Chinese medicine, has been utilized for several decades in the treatment of hepatocellular carcinoma (HCC). Previous research has demonstrated its anti-tumour efficacy, but its underlying mechanism remains unclear. This study aimed to assess the therapeutic effects of luteolin in H22 tumour-bearing mice. luteolin effectively inhibited the growth of solid tumours in a well-established mouse model of HCC. High-throughput sequencing revealed that luteolin treatment could enhance T-cell activation, cell chemotaxis and cytokine production. In addition, luteolin helped sustain a high ratio of CD8+ T lymphocytes in the spleen, peripheral blood and tumour tissues. The effects of luteolin on the phenotypic and functional changes in tumour-infiltrating CD8+ T lymphocytes were also investigated. Luteolin restored the cytotoxicity of tumour-infiltrating CD8+ T lymphocytes in H22 tumour-bearing mice. The CD8+ T lymphocytes exhibited intensified phenotype activation and increased production of granzyme B, IFN-γ and TNF-α in serum. The combined administration of luteolin and the PD-1 inhibitor enhanced the anti-tumour effects in H22 tumour-bearing mice. Luteolin could exert an anti-tumour immune response by inducing CD8+ T lymphocyte infiltration and enhance the anti-tumour effects of the PD-1 inhibitor on H22 tumour-bearing mice.


Subject(s)
CD8-Positive T-Lymphocytes , Carcinoma, Hepatocellular , Liver Neoplasms , Luteolin , Lymphocytes, Tumor-Infiltrating , Luteolin/pharmacology , Luteolin/therapeutic use , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Mice , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/metabolism , Cell Line, Tumor , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Cytokines/metabolism , Male , Granzymes/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Mice, Inbred C57BL
7.
BMC Pediatr ; 24(1): 587, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39285316

ABSTRACT

BACKGROUND: Congenital cytomegalovirus (cCMV) infection can lead to a range of adverse outcomes. The majority of cCMV neonates with clinical symptoms are infected postnatally; however, established cases of intrauterine infection are uncommon, resulting in a paucity of reports on clinical findings and lymphocytes expression in CMV-infected neonates. CASE PRESENTATION: We followed a neonate with cCMV infection from the onset of hospitalization to several months of follow-up. This infant was intrauterine CMV-positive in the amniotic fluid of the mother at 21 weeks' gestation and received intravenous ganciclovir infusion and sequential oral valganciclovir after birth. The typical clinical signs manifested in the nervous system, liver, and peripheral blood and were documented during the hospitalizaion period and up to the follow-up visit. Flow cytometry was employed to examine the expression of T cells, their subsets, and the associated cytokines in peripheral blood samples at various time points. The flow data for the cCMV neonate were compared with those of the controls at each time point. Following treatment, clinical symptoms improved and the infant became CMV negative. However, developmental delays occurred later in life. The proportion of CD8+CD28- Tregs in the peripheral blood of the neonate with congenital CMV infection was higher than that in the controls at the three time points. The expression levels of perforin and granzyme B secreted by γδ T cells (Vδ1 and Vδ2 T cells), increased during the course of hospitalization until follow-up and were higher than those in the controls at the three time points. CONCLUSIONS: Despite the alleviation of clinical symptoms, developmental delay in later life remains inevitable in this intrauterine cCMV neonate. CD8+CD28- Tregs and Vδ1 and Vδ2 T cells secreting perforin and granzyme B may be involved in congenital CMV infection, although this hypothesis requires validation in a larger study. This report may contribute to our understanding of the effect of current treatment and the immune status of intrauterine cCMV-infected neonates.


Subject(s)
Cytomegalovirus Infections , T-Lymphocytes, Regulatory , Humans , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/diagnosis , Cytomegalovirus Infections/drug therapy , Infant, Newborn , Female , T-Lymphocytes, Regulatory/immunology , Pregnancy , CD8-Positive T-Lymphocytes/immunology , CD28 Antigens , Pregnancy Complications, Infectious , Perforin/metabolism , Antiviral Agents/therapeutic use , Male , Ganciclovir/therapeutic use , Granzymes/metabolism
8.
Signal Transduct Target Ther ; 9(1): 245, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39300122

ABSTRACT

Pyroptosis is a type of programmed cell death characterized by cell swelling and osmotic lysis, resulting in cytomembrane rupture and release of immunostimulatory components, which play a role in several pathological processes. Significant cellular responses to various stimuli involve the formation of inflammasomes, maturation of inflammatory caspases, and caspase-mediated cleavage of gasdermin. The function of pyroptosis in disease is complex but not a simple angelic or demonic role. While inflammatory diseases such as sepsis are associated with uncontrollable pyroptosis, the potent immune response induced by pyroptosis can be exploited as a therapeutic target for anti-tumor therapy. Thus, a comprehensive review of the role of pyroptosis in disease is crucial for further research and clinical translation from bench to bedside. In this review, we summarize the recent advancements in understanding the role of pyroptosis in disease, covering the related development history, molecular mechanisms including canonical, non-canonical, caspase 3/8, and granzyme-mediated pathways, and its regulatory function in health and multiple diseases. Moreover, this review also provides updates on promising therapeutic strategies by applying novel small molecule inhibitors and traditional medicines to regulate pyroptosis. The present dilemmas and future directions in the landscape of pyroptosis are also discussed from a clinical perspective, providing clues for scientists to develop novel drugs targeting pyroptosis.


Subject(s)
Pyroptosis , Pyroptosis/genetics , Humans , Inflammasomes/metabolism , Inflammasomes/genetics , Inflammasomes/immunology , Granzymes/genetics , Granzymes/metabolism , Sepsis/genetics , Sepsis/pathology , Sepsis/metabolism , Sepsis/immunology , Caspase 8/genetics , Caspase 8/metabolism , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/metabolism , Neoplasms/immunology , Neoplasms/drug therapy , Signal Transduction
9.
Biochem J ; 481(18): 1255-1274, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39248243

ABSTRACT

Tauopathies, including Alzheimer's disease, corticobasal degeneration and progressive supranuclear palsy, are characterised by the aggregation of tau into insoluble neurofibrillary tangles in the brain. Tau is subject to a range of post-translational modifications, including proteolysis, that can promote its aggregation. Neuroinflammation is a hallmark of tauopathies and evidence is growing for a role of CD8+ T cells in disease pathogenesis. CD8+ T cells release granzyme proteases but what role these proteases play in neuronal dysfunction is currently lacking. Here, we identified that granzyme A (GzmA) is present in brain tissue and proteolytically cleaves tau. Mass spectrometric analysis of tau fragments produced on digestion of tau with GzmA identified three cleavage sites at R194-S195, R209-S210 and K240-S241. Mutation of the critical Arg or Lys residues at the cleavage sites in tau or chemical inhibition of GzmA blocked the proteolysis of tau by GzmA. Development of a semi-targeted mass spectrometry approach identified peptides in tauopathy brain tissue corresponding to proteolysis by GzmA at R209-S210 and K240-S241 in tau. When expressed in cells the GzmA-cleaved C-terminal fragments of tau were highly phosphorylated and aggregated upon incubation of the cells with tauopathy brain seed. The C-terminal fragment tau195-441 was able to transfer between cells and promote aggregation of tau in acceptor cells, indicating the propensity for such tau fragments to propagate between cells. Collectively, these results raise the possibility that GzmA, released from infiltrating cytotoxic CD8+ T cells, proteolytically cleaves tau into fragments that may contribute to its pathological properties in tauopathies.


Subject(s)
Granzymes , Proteolysis , Tauopathies , tau Proteins , Humans , tau Proteins/metabolism , tau Proteins/genetics , Granzymes/metabolism , Granzymes/genetics , Tauopathies/metabolism , Tauopathies/pathology , Tauopathies/genetics , Brain/metabolism , Brain/pathology , CD8-Positive T-Lymphocytes/metabolism , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/genetics
10.
Exp Dermatol ; 33(9): e15172, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39219105

ABSTRACT

The pemphigoid disease epidermolysis bullosa acquisita (EBA) is an autoimmune blistering skin disease characterized by autoantibodies against type VII collagen (COL7), immune cell infiltrates at the dermal-epidermal junction and subepidermal blistering. Proteases, particularly granzyme B (GzmB), have been established as therapeutic targets for the treatment of EBA and other pemphigoid diseases. We investigated the impact of the novel GzmB inhibitor SNT-6935 on anti-COL7 IgG-induced subepidermal blistering in a well-established EBA ex vivo model. Our findings demonstrate that pharmacological targeting of GzmB with its selective inhibitor SNT-6935 significantly reduced autoantibody-induced dermal-epidermal separation in human skin cryosections. Interestingly, treatment of skin cryosections with recombinant human GzmB alone did not cause dermal-epidermal separation, suggesting that additional mechanisms alongside GzmB are required for tissue damage in EBA. In conclusion, our study highlights the significant contribution of GzmB to the pathogenesis of EBA and supports the notion of GzmB as a therapeutic target in EBA and other pemphigoid diseases.


Subject(s)
Autoantibodies , Collagen Type VII , Epidermis , Epidermolysis Bullosa Acquisita , Granzymes , Humans , Collagen Type VII/immunology , Dermis/pathology , Epidermis/pathology , Epidermolysis Bullosa Acquisita/drug therapy , Epidermolysis Bullosa Acquisita/immunology , Granzymes/metabolism , Granzymes/antagonists & inhibitors , Skin/pathology
11.
Sci Adv ; 10(34): eadp2636, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39178263

ABSTRACT

SARS-CoV-2 infection induces the generation of virus-specific CD4+ and CD8+ effector and memory T cells. However, the contribution of T cells in controlling SARS-CoV-2 during infection is not well understood. Following infection of C57BL/6 mice, SARS-CoV-2-specific CD4+ and CD8+ T cells are recruited to the respiratory tract, and a vast proportion secrete the cytotoxic molecule granzyme B. Using depleting antibodies, we found that T cells within the lungs play a minimal role in viral control, and viral clearance occurs in the absence of both CD4+ and CD8+ T cells through 28 days postinfection. In the nasal compartment, depletion of both CD4+ and CD8+ T cells, but not individually, results in persistent, culturable virus replicating in the nasal epithelial layer through 28 days postinfection. Viral sequencing analysis revealed adapted mutations across the SARS-CoV-2 genome, including a large deletion in ORF6. Overall, our findings highlight the importance of T cells in controlling virus replication within the respiratory tract during SARS-CoV-2 infection.


Subject(s)
CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , COVID-19 , Mice, Inbred C57BL , SARS-CoV-2 , Virus Replication , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , SARS-CoV-2/physiology , SARS-CoV-2/immunology , COVID-19/virology , COVID-19/immunology , COVID-19/prevention & control , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , CD4-Positive T-Lymphocytes/metabolism , Mice , Lung/virology , Lung/immunology , Humans , Female , Nasal Mucosa/virology , Nasal Mucosa/immunology , Nasal Mucosa/metabolism , Granzymes/metabolism
12.
Nat Cardiovasc Res ; 3(8): 970-986, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39196030

ABSTRACT

Doxorubicin, the most prescribed chemotherapeutic drug, causes dose-dependent cardiotoxicity and heart failure. However, our understanding of the immune response elicited by doxorubicin is limited. Here we show that an aberrant CD8+ T cell immune response following doxorubicin-induced cardiac injury drives adverse remodeling and cardiomyopathy. Doxorubicin treatment in non-tumor-bearing mice increased circulating and cardiac IFNγ+CD8+ T cells and activated effector CD8+ T cells in lymphoid tissues. Moreover, doxorubicin promoted cardiac CD8+ T cell infiltration and depletion of CD8+ T cells in doxorubicin-treated mice decreased cardiac fibrosis and improved systolic function. Doxorubicin treatment induced ICAM-1 expression by cardiac fibroblasts resulting in enhanced CD8+ T cell adhesion and transformation, contact-dependent CD8+ degranulation and release of granzyme B. Canine lymphoma patients and human patients with hematopoietic malignancies showed increased circulating CD8+ T cells after doxorubicin treatment. In human cancer patients, T cells expressed IFNγ and CXCR3, and plasma levels of the CXCR3 ligands CXCL9 and CXCL10 correlated with decreased systolic function.


Subject(s)
Disease Models, Animal , Doxorubicin , Fibrosis , Interferon-gamma , T-Lymphocytes, Cytotoxic , Animals , Doxorubicin/adverse effects , Fibrosis/chemically induced , Humans , Dogs , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Interferon-gamma/metabolism , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/toxicity , Mice, Inbred C57BL , Cardiotoxicity/etiology , Receptors, CXCR3/metabolism , Chemokine CXCL10/metabolism , Male , Granzymes/metabolism , Cardiomyopathies/chemically induced , Cardiomyopathies/pathology , Cardiomyopathies/immunology , Myocardium/pathology , Myocardium/metabolism , Myocardium/immunology , Cell Degranulation/drug effects , Chemokine CXCL9/metabolism , Ventricular Function, Left/drug effects , Systole/drug effects , Mice , Female , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Cell Adhesion/drug effects , Lymphocyte Activation/drug effects
13.
Cells ; 13(16)2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39195200

ABSTRACT

BACKGROUND: Large granular lymphocyte leukemias (LGLLs) are rare lymphoproliferative malignancies caused by clonal expansion of granular lymphocytes. T-cell LGLL and natural killer (NK) cell LGLL are defined based on their cellular origin. Their clinical manifestation and pathophysiology vary depending on the subtype and include, e.g., neutropenia, anemia, recurrent infections, and autoimmunity. A limited number of available patient-derived cell lines are considered valuable tools to study the biology of these malignancies. They differ in the expression of lineage-specific surface markers, but generally contain cytotoxic effector molecules in characteristic granules. METHODS: We investigated the presence and release of lysosome-associated effector proteins in patient-derived LGLL cell lines by flow and imaging cytometry, by Western blotting and by bottom-up proteomics profiling. RESULTS: The tested cell lines did not express FasL (CD178), but did express CD26/DPP4+. Intracellularly, we detected major differences in the abundance and subcellular distribution of granzymes, perforin, and granulysin. Similar differences were seen in enriched lysosome-related effector vesicles (LREVs). The proteomics profiling of enriched EVs from an NK-LGLL line (NKL) and a T-LGLL line (MOTN-1), confirmed individual profiles of effector molecules. CONCLUSION: Our analyses underscore the individual distribution of effector proteins but also open new routes to define the role of intra- and extracellular granules in the disease manifestation or pathology of LGLLs.


Subject(s)
Extracellular Vesicles , Leukemia, Large Granular Lymphocytic , Humans , Leukemia, Large Granular Lymphocytic/pathology , Leukemia, Large Granular Lymphocytic/metabolism , Extracellular Vesicles/metabolism , Cell Line, Tumor , Cytoplasmic Granules/metabolism , Lysosomes/metabolism , Proteomics , Killer Cells, Natural/metabolism , Perforin/metabolism , Granzymes/metabolism , Antigens, Differentiation, T-Lymphocyte
14.
Elife ; 132024 Aug 12.
Article in English | MEDLINE | ID: mdl-39133873

ABSTRACT

Group 1 innate lymphoid cells (ILCs) comprise conventional natural killer (cNK) cells and type 1 innate lymphoid cells (ILC1s). The main functions of liver cNK cells and ILC1s not only include directly killing target cells but also regulating local immune microenvironment of the liver through the secretion of cytokines. Uncovering the intricate mechanisms by which transcriptional factors regulate and influence the functions of liver cNK cells and ILC1s, particularly within the context of liver tumors, presents a significant opportunity to amplify the effectiveness of immunotherapies against liver malignancies. Using Ncr1-drived conditional knockout mouse model, our study reveals the regulatory role of Prdm1 in shaping the composition and maturation of cNK cells. Although Prdm1 did not affect the killing function of cNK cells in an in vivo cytotoxicity model, a significant increase in cancer metastasis was observed in Prdm1 knockout mice. Interferon-gamma (IFN-γ), granzyme B, and perforin secretion decreased significantly in Prdm1-deficient cNK cells and liver ILC1s. Single-cell RNA sequencing (scRNA-seq) data also provided evidences that Prdm1 maintains functional subsets of cNK cells and liver ILC1s and facilitates communications between cNK cells, liver ILC1s, and macrophages. The present study unveiled a novel regulatory mechanism of Prdm1 in cNK cells and liver ILC1s, showing promising potential for developing innovative immune therapy strategies against liver cancer.


Subject(s)
Liver Neoplasms , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1 , Animals , Mice , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Liver Neoplasms/immunology , Liver Neoplasms/genetics , Killer Cells, Natural/immunology , Interferon-gamma/metabolism , Immunity, Innate , Lymphocytes/immunology , Immunologic Surveillance , Granzymes/metabolism , Granzymes/genetics , Natural Cytotoxicity Triggering Receptor 1/metabolism , Natural Cytotoxicity Triggering Receptor 1/genetics , Perforin/metabolism , Perforin/genetics , Liver/immunology , Liver/metabolism , Mice, Inbred C57BL , Tumor Microenvironment/immunology , Antigens, Ly
15.
Fish Shellfish Immunol ; 153: 109865, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39214265

ABSTRACT

Secreted by natural killer cells and cytotoxic T lymphocytes, Granzyme B is involved in regulating the adaptive immune response in vertebrates and plays a pivotal role in resisting virus invasion and removing pathogens. Although it had been extensively studied in mammals, the involvement of Granzyme B in adaptive immune response of early vertebrates remained elusive. In this study, we investigated the Granzyme B in Oreochromis niloticus (OnGrB), found that its function domain was conserved. Additionally, OnGrB was widely expressed in various tissues and could respond to T-cell activation in vitro at the transcriptional level. Furthermore, we prepared the recombinant OnGrB (rOnGrB) as an immunogen to develop a mouse anti-OnGrB monoclonal antibody (mAb). Using this anti-OnGrB mAb as a tool, we explored the expression of OnGrB in the adaptive immune response of tilapia. Our findings revealed that T cell was a significant source of OnGrB production, the expression of OnGrB at the protein level and the proportion of OnGrB + T cells increased after both T cell activation in vitro and infection with Edwardsiella piscicida in vivo. More importantly, our findings also preliminarily illuminated that p65 could regulate the transcriptional activity of OnGrB. These results indicated that OnGrB was involved in the adaptive immunity of tilapia and played a critical role in T cell function in teleost. Our study provided theoretical support and new perspectives for understanding adaptive immunity in teleost.


Subject(s)
Cichlids , Edwardsiella , Enterobacteriaceae Infections , Fish Diseases , Fish Proteins , Granzymes , Animals , Adaptive Immunity , Amino Acid Sequence , Cichlids/immunology , Cichlids/genetics , Edwardsiella/immunology , Edwardsiella/physiology , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/veterinary , Fish Diseases/immunology , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/veterinary , Gene Expression Regulation/immunology , Granzymes/genetics , Granzymes/immunology , Granzymes/metabolism , Phylogeny , Sequence Alignment/veterinary , T-Lymphocytes/immunology
16.
Int J Mol Sci ; 25(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39125711

ABSTRACT

Cystatin F (CstF) is a protease inhibitor of cysteine cathepsins, including those involved in activating the perforin/granzyme cytotoxic pathways. It is targeted at the endolysosomal pathway but can also be secreted to the extracellular milieu or endocytosed by bystander cells. CstF was shown to be significantly increased in tuberculous pleurisy, and during HIV coinfection, pleural fluids display high viral loads. In human macrophages, our previous results revealed a strong upregulation of CstF in phagocytes activated by interferon γ or after infection with Mycobacterium tuberculosis (Mtb). CstF manipulation using RNA silencing led to increased proteolytic activity of lysosomal cathepsins, improving Mtb intracellular killing. In the present work, we investigate the impact of CstF depletion in macrophages during the coinfection of Mtb-infected phagocytes with lymphocytes infected with HIV. The results indicate that decreasing the CstF released by phagocytes increases the major pro-granzyme convertase cathepsin C of cytotoxic immune cells from peripheral blood-derived lymphocytes. Consequently, an observed augmentation of the granzyme B cytolytic activity leads to a significant reduction in viral replication in HIV-infected CD4+ T-lymphocytes. Ultimately, this knowledge can be crucial for developing new therapeutic approaches to control both pathogens based on manipulating CstF.


Subject(s)
Cathepsin C , Coinfection , Granzymes , HIV Infections , Macrophages , Mycobacterium tuberculosis , Humans , Granzymes/metabolism , Granzymes/genetics , HIV Infections/metabolism , HIV Infections/immunology , Macrophages/metabolism , Macrophages/immunology , Macrophages/microbiology , Macrophages/virology , Coinfection/microbiology , Cathepsin C/metabolism , Cathepsin C/genetics , Cystatins/metabolism , Cystatins/genetics , Tuberculosis/metabolism , Tuberculosis/immunology , Tuberculosis/microbiology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , HIV-1/physiology , Biomarkers, Tumor
17.
Int J Mol Sci ; 25(16)2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39201366

ABSTRACT

Pterygium is often associated with chronic ultraviolet (UV) radiation exposure and characterized by the overgrowth of conjunctiva and extracellular matrix (ECM) remodeling. Notably, several studies in the skin have demonstrated that chronic UV radiation can upregulate Granzyme B (GrB) expression and increase ECM degradation. The aim of this study was to compare GrB expression between pterygium and healthy controls and to further link this GrB expression to mast cells. Post-mortem pterygium tissues and conjunctival tissues from age-matched controls were used to assess GrB expression via immunofluorescence and microscopy. We found a significantly higher density of GrB+ cells from pterygium specimens compared to healthy controls. Furthermore, many of the GrB+ cells in pterygium specimens co-expressed tryptase, a mast cell marker. These findings suggest a role for conjunctival mast cell-secreted GrB in the pathogenesis of pterygium and highlight GrB as a possible therapeutic target in delaying or halting pterygium progression.


Subject(s)
Conjunctiva , Granzymes , Pterygium , Humans , Pterygium/metabolism , Pterygium/pathology , Granzymes/metabolism , Conjunctiva/metabolism , Conjunctiva/pathology , Male , Female , Middle Aged , Aged , Mast Cells/metabolism , Adult , Case-Control Studies , Aged, 80 and over , Tryptases/metabolism
18.
Genes Genomics ; 46(9): 1097-1106, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39115674

ABSTRACT

BACKGROUND: Granzymes are essential serine proteases in cytotoxic T cells and natural killer (NK) cells, with GZMK's expression being less understood. This study aims to uncover GZMK expression profiles across various immune cell types using single-cell RNA sequencing meta-analysis. OBJECTIVE: This study aims to uncover GZMK expression profiles across various immune cell types using single-cell RNA sequencing meta-analysis. METHODS: We conducted a meta-analysis using cellxgene, an interactive data exploration platform developed by the Chan Zuckerberg Initiative. We focused on mature T cells, NK cells, B cells, and NKT cells. We also checked transcription factor binding sites at the granzyme gene promoter regions using JASPAR. Comparative analysis was also done using mouse single-cell RNA sequencing data. RESULTS: GZMK was the most lowly expressed in NK cells and mature NKT cells in most tissues except for colon and lymph nodes. In mature T cells, GZMK is similarly or more highly expressed than other granzymes. HBCA data revealed weak expression of GZMK in NK cells but strong expression in effector memory CD8-positive, alpha-beta T cells. Combined data shows no significant difference in GZMK expression between cell types. Subtype analysis shows that GZMK expression was higher in CD16-negative, CD56-bright NK cells when compared to CD16-positive, CD56-dim NK cells. We also identified unique transcription factor binding sites for GZMK. While this pattern in mouse data with low Gzmk expression in NK cells and higher T cells was repeated. CONCLUSION: GZMK expression is distinctively regulated among immune cells and tissues, with unique promoter regions and transcription factor binding sites contributing to this differential expression. These insights into GZMK's role in immune function and regulation offer potential therapeutic targets.


Subject(s)
Granzymes , Killer Cells, Natural , Single-Cell Analysis , Granzymes/genetics , Granzymes/metabolism , Animals , Single-Cell Analysis/methods , Mice , Killer Cells, Natural/metabolism , Killer Cells, Natural/immunology , Humans , RNA-Seq/methods , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Natural Killer T-Cells/metabolism , Natural Killer T-Cells/immunology , Promoter Regions, Genetic , Binding Sites , Single-Cell Gene Expression Analysis
19.
Mol Immunol ; 174: 1-10, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39126837

ABSTRACT

BACKGROUND: Cannabidiol (CBD), the major non-psychoactive component of cannabis, exhibits anti-inflammatory properties, but less is known about the immunomodulatory potential of CBD on activated natural killer (NK) cells and/or their targets. Many tumor cells present heat shock protein 70 (Hsp70) on their cell surface in a tumor-specific manner and although a membrane Hsp70 (mHsp70) positive phenotype serves as a target for Hsp70-activated NK cells, a high mHsp70 expression is associated with tumor aggressiveness. This study investigated the immuno-modulatory potential of CBD on NK cells stimulated with TKD Hsp70 peptide and IL-2 (TKD+IL-2) and also on HCT116 p53wt and HCT116 p53-/- colorectal cancer cells exhibiting high and low basal levels of mHsp70 expression. RESULTS: Apart from an increase in the density of NTB-A and a reduced expression of LAMP-1, the expression of all other activatory NK cell receptors including NKp30, NKG2D and CD69 which are significantly up-regulated after stimulation with TKD+IL-2 remained unaffected after a co-treatment with CBD. However, the release of major pro-inflammatory cytokines by NK cells such as interferon-γ (IFN-γ) and the effector molecule granzyme B (GrzB) was significantly reduced upon CBD treatment. With respect to the tumor target cells, CBD significantly reduced the elevated expression of mHsp70 but had no effect on the low basal mHsp70 expression. Expression of other NK cell ligands such as MICA and MICB remained unaffected, and the NK cell ligands ULBP and B7-H6 were not expressed on these target cells. Consistent with the reduced mHsp70 expression, treatment of both effector and target cells with CBD reduced the killing of high mHsp70 expressing tumor cells by TKD+IL-2+CBD pre-treated NK cells but had no effect on the killing of low mHsp70 expressing tumor cells. Concomitantly, CBD treatment reduced the TKD+IL-2 induced increased release of IFN-γ, IL-4, TNF-α and GrzB, but CBD had no effect on the release of IFN-α when NK cells were co-incubated with tumor target cells. CONCLUSION: Cannabidiol (CBD) may potentially diminish the anti-tumor effectiveness of TKD+IL-2 activated natural killer (NK) cells.


Subject(s)
Cannabidiol , HSP70 Heat-Shock Proteins , Killer Cells, Natural , Lymphocyte Activation , Humans , Cannabidiol/pharmacology , Killer Cells, Natural/immunology , Killer Cells, Natural/drug effects , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , HSP70 Heat-Shock Proteins/metabolism , HCT116 Cells , Immunologic Factors/pharmacology , Interleukin-2/metabolism , Interleukin-2/immunology , Granzymes/metabolism , Interferon-gamma/metabolism , Interferon-gamma/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects
20.
Int J Infect Dis ; 147: 107206, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39147194

ABSTRACT

BACKGROUND: Cutaneous leishmaniasis (CL) caused by Leishmania (Viannia) braziliensis is associated with an inflammatory response. Granzyme (GzmB) and IL-1ß play a key role in the pathology. Meglumine antimoniate (MA) is the first-choice drug for the treatment of CL, but therapy failure is observed in up to 50% of the cases. The protein, rSm29 of Schistosoma mansoni, down-modulates pro-inflammatory cytokine production. We evaluate if the combination of topical rSm29 plus MA increases the cure rate of CL. METHODS: In this randomized clinical trial, 91 CL patients were allocated in 3 groups. All cases received MA (20 mg/kg/weight) for 20 days. Group 1 used topical rSm29 (10 µg), group 2 a placebo topically applied, and group 3 received only MA. RESULTS: The cure rate on day 90 was 71% in subjects treated with rSm29 plus MA, and 43% in patients who received MA plus placebo or MA alone (P < 0.05). There was a decrease in GzmB and an increase in IFN-γ (P < 0.05) in supernatants of skin biopsies of the lesions obtained on D7 of therapy (P < 0.05) in patients who received rSm29. CONCLUSION: rSm29 associated with MA reduces GzmB levels, is more effective than MA alone, and decreases CL healing time. CLINICAL TRIALS REGISTRATION: ClinicalTrial.gov under NCT06000514.


Subject(s)
Administration, Topical , Antiprotozoal Agents , Drug Therapy, Combination , Leishmaniasis, Cutaneous , Meglumine Antimoniate , Organometallic Compounds , Humans , Leishmaniasis, Cutaneous/drug therapy , Leishmaniasis, Cutaneous/parasitology , Meglumine Antimoniate/therapeutic use , Meglumine Antimoniate/administration & dosage , Male , Female , Adult , Antiprotozoal Agents/therapeutic use , Antiprotozoal Agents/administration & dosage , Middle Aged , Young Adult , Organometallic Compounds/therapeutic use , Organometallic Compounds/administration & dosage , Treatment Outcome , Meglumine/administration & dosage , Meglumine/therapeutic use , Adolescent , Animals , Leishmania braziliensis/drug effects , Administration, Intravenous , Granzymes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL