Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51.317
Filter
1.
Front Immunol ; 15: 1414298, 2024.
Article in English | MEDLINE | ID: mdl-38938577

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is one of the most common tumor entities worldwide, with human papillomavirus (HPV) infection contributing to cancer development. Conventional therapies achieve only limited efficiency, especially in recurrent or metastatic HNSCC. As the immune landscape decisively impacts the survival of patients and treatment efficacy, this study comprehensively investigated the immunological tumor microenvironment (TME) and its association with patient outcome, with special focus on several dendritic cell (DC) and T lymphocyte subpopulations. Therefore, formalin-fixed paraffin-embedded tumor samples of 56 HNSCC patients, who have undergone resection and adjuvant radiotherapy, were analyzed by multiplex immunohistochemistry focusing on the detailed phenotypic characterization and spatial distribution of DCs, CD8+ T cells, and T-helper cell subsets in different tumor compartments. Immune cell densities and proportions were correlated with clinical characteristics of the whole HNSCC cohort and different HPV- or hypoxia-associated subcohorts. Tumor stroma was highly infiltrated by plasmacytoid DCs and T lymphocytes. Among the T-helper cells and CD8+ T cells, stromal regulatory T cells and intraepithelial exhausted CD8+ T cells expressing programmed cell death protein-1 (PD-1+) and/or lymphocyte-activation gene-3 (LAG-3+) were the predominant phenotypes, indicating an immunosuppressive TME. HPV-associated tumors showed significantly higher infiltration of type I and type II conventional DCs (cDC1, cDC2) as well as several CD8+ T cell phenotypes including exhausted, activated, and proliferating T cells. On the contrary, tumors with hypoxia-associated gene signatures exhibited reduced infiltration for these immune cells. By multivariate Cox regression, immune-related prognostic factors were identified. Patient clusters defined by high infiltration of DCs and T lymphocytes combined with HPV positivity or low hypoxia showed significantly prolonged survival. Thereby, cDC1 and CD8+ T cells emerged as independent prognostic factors for local and distant recurrence. These results might contribute to the implementation of an immune cell infiltration score predicting HNSCC patients' survival and such patient stratification might improve the design of future individualized radiochemo-(immuno)therapies.


Subject(s)
CD8-Positive T-Lymphocytes , Dendritic Cells , Head and Neck Neoplasms , Squamous Cell Carcinoma of Head and Neck , Tumor Microenvironment , Humans , Dendritic Cells/immunology , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/virology , Squamous Cell Carcinoma of Head and Neck/mortality , Male , Female , CD8-Positive T-Lymphocytes/immunology , Middle Aged , Tumor Microenvironment/immunology , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/virology , Head and Neck Neoplasms/mortality , Aged , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Prognosis , Adult , Papillomavirus Infections/immunology , Papillomavirus Infections/complications , Papillomavirus Infections/virology
2.
Science ; 384(6703): eadh4567, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38935717

ABSTRACT

Inflammation and tissue damage associated with pancreatitis can precede or occur concurrently with pancreatic ductal adenocarcinoma (PDAC). We demonstrate that in PDAC coupled with pancreatitis (ptPDAC), antigen-presenting type I conventional dendritic cells (cDC1s) are specifically activated. Immune checkpoint blockade therapy (iCBT) leads to cytotoxic CD8+ T cell activation and elimination of ptPDAC with restoration of life span even upon PDAC rechallenge. Using PDAC antigen-loaded cDC1s as a vaccine, immunotherapy-resistant PDAC was rendered sensitive to iCBT with elimination of tumors. cDC1 vaccination coupled with iCBT identified specific CDR3 sequences in the tumor-infiltrating CD8+ T cells with potential therapeutic importance. This study identifies a fundamental difference in the immune microenvironment in PDAC concurrent with, or without, pancreatitis and provides a rationale for combining cDC1 vaccination with iCBT as a potential treatment option.


Subject(s)
Carcinoma, Pancreatic Ductal , Dendritic Cells , Immunotherapy , Pancreatic Neoplasms , Tumor Microenvironment , Dendritic Cells/immunology , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/immunology , Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/immunology , Immunotherapy/methods , Animals , Mice , Humans , Tumor Microenvironment/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Pancreatitis/immunology , Pancreatitis/therapy , CD8-Positive T-Lymphocytes/immunology , Mice, Inbred C57BL
3.
Curr Oncol ; 31(6): 3099-3121, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38920720

ABSTRACT

Epithelial ovarian cancer (EOC) has not significantly benefited from advances in immunotherapy, mainly because of the lack of well-defined actionable antigen targets. Using proteogenomic analyses of primary EOC tumors, we previously identified 91 aberrantly expressed tumor-specific antigens (TSAs) originating from unmutated genomic sequences. Most of these TSAs derive from non-exonic regions, and their expression results from cancer-specific epigenetic changes. The present study aimed to evaluate the immunogenicity of 48 TSAs selected according to two criteria: presentation by highly prevalent HLA allotypes and expression in a significant fraction of EOC tumors. Using targeted mass spectrometry analyses, we found that pulsing with synthetic TSA peptides leads to a high-level presentation on dendritic cells. TSA abundance correlated with the predicted binding affinity to the HLA allotype. We stimulated naïve CD8 T cells from healthy blood donors with TSA-pulsed dendritic cells and assessed their expansion with two assays: MHC-peptide tetramer staining and TCR Vß CDR3 sequencing. We report that these TSAs can expand sizeable populations of CD8 T cells and, therefore, represent attractive targets for EOC immunotherapy.


Subject(s)
Antigens, Neoplasm , Ovarian Neoplasms , Humans , Female , Antigens, Neoplasm/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/genetics , Dendritic Cells/immunology , Carcinoma, Ovarian Epithelial/immunology , Carcinoma, Ovarian Epithelial/genetics , CD8-Positive T-Lymphocytes/immunology , Immunotherapy/methods
4.
Nat Commun ; 15(1): 5413, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926424

ABSTRACT

Diet composition impacts metabolic health and is now recognized to shape the immune system, especially in the intestinal tract. Nutritional imbalance and increased caloric intake are induced by high-fat diet (HFD) in which lipids are enriched at the expense of dietary fibers. Such nutritional challenge alters glucose homeostasis as well as intestinal immunity. Here, we observed that short-term HFD induced dysbiosis, glucose intolerance and decreased intestinal RORγt+ CD4 T cells, including peripherally-induced Tregs and IL17-producing (Th17) T cells. However, supplementation of HFD-fed male mice with the fermentable dietary fiber fructooligosaccharides (FOS) was sufficient to maintain RORγt+ CD4 T cell subsets and microbial species known to induce them, alongside having a beneficial impact on glucose tolerance. FOS-mediated normalization of Th17 cells and amelioration of glucose handling required the cDC2 dendritic cell subset in HFD-fed animals, while IL-17 neutralization limited FOS impact on glucose tolerance. Overall, we uncover a pivotal role of cDC2 in the control of the immune and metabolic effects of FOS in the context of HFD feeding.


Subject(s)
Dendritic Cells , Diet, High-Fat , Homeostasis , Mice, Inbred C57BL , Oligosaccharides , Animals , Oligosaccharides/pharmacology , Diet, High-Fat/adverse effects , Dendritic Cells/immunology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Male , Mice , Th17 Cells/immunology , Th17 Cells/metabolism , Th17 Cells/drug effects , Glucose/metabolism , Interleukin-17/metabolism , Dietary Fiber/pharmacology , Glucose Intolerance/immunology , Glucose Intolerance/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Dysbiosis/immunology , Gastrointestinal Microbiome/drug effects
5.
Viruses ; 16(6)2024 May 29.
Article in English | MEDLINE | ID: mdl-38932162

ABSTRACT

Vaccinia virus is the most successful vaccine in human history and functions as a protective vaccine against smallpox and monkeypox, highlighting the importance of ongoing research into vaccinia due to its genetic similarity to other emergent poxviruses. Moreover, vaccinia's ability to accommodate large genetic insertions makes it promising for vaccine development and potential therapeutic applications, such as oncolytic agents. Thus, understanding how superior immunity is generated by vaccinia is crucial for designing other effective and safe vaccine strategies. During vaccinia inoculation by scarification, the skin serves as a primary site for the virus-host interaction, with various cell types playing distinct roles. During this process, hematopoietic cells undergo abortive infections, while non-hematopoietic cells support the full viral life cycle. This differential permissiveness to viral replication influences subsequent innate and adaptive immune responses. Dendritic cells (DCs), key immune sentinels in peripheral tissues such as skin, are pivotal in generating T cell memory during vaccinia immunization. DCs residing in the skin capture viral antigens and migrate to the draining lymph nodes (dLN), where they undergo maturation and present processed antigens to T cells. Notably, CD8+ T cells are particularly significant in viral clearance and the establishment of long-term protective immunity. Here, we will discuss vaccinia virus, its continued relevance to public health, and viral strategies permissive to immune escape. We will also discuss key events and populations leading to long-term protective immunity and remaining key gaps.


Subject(s)
Immune Evasion , Vaccinia virus , Vaccinia , Vaccinia virus/immunology , Vaccinia virus/genetics , Humans , Animals , Vaccinia/immunology , Vaccinia/virology , Dendritic Cells/immunology , Virus Replication , Adaptive Immunity , CD8-Positive T-Lymphocytes/immunology
6.
Viruses ; 16(6)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38932202

ABSTRACT

Previous studies from our laboratory and others have established the dendritic cell (DC) as a key target of RSV that drives infection-induced pathology. Analysis of RSV-induced transcriptomic changes in RSV-infected DC revealed metabolic gene signatures suggestive of altered cellular metabolism. Reverse phase protein array (RPPA) data showed significantly increased PARP1 phosphorylation in RSV-infected DC. Real-time cell metabolic analysis demonstrated increased glycolysis in PARP1-/- DC after RSV infection, confirming a role for PARP1 in regulating DC metabolism. Our data show that enzymatic inhibition or genomic ablation of PARP1 resulted in increased ifnb1, il12, and il27 in RSV-infected DC which, together, promote a more appropriate anti-viral environment. PARP1-/- mice and PARP1-inhibitor-treated mice were protected against RSV-induced immunopathology including airway inflammation, Th2 cytokine production, and mucus hypersecretion. However, delayed treatment with PARP1 inhibitor in RSV-infected mice provided only partial protection, suggesting that PARP1 is most important during the earlier innate immune stage of RSV infection.


Subject(s)
Dendritic Cells , Lung , Poly (ADP-Ribose) Polymerase-1 , Respiratory Syncytial Virus Infections , Respiratory Syncytial Viruses , Animals , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , Mice , Dendritic Cells/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Lung/immunology , Lung/pathology , Lung/virology , Respiratory Syncytial Viruses/immunology , Mice, Knockout , Cytokines/metabolism , Cytokines/immunology , Immunity, Innate , Female
7.
Lancet Oncol ; 25(7): 865-878, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38848742

ABSTRACT

BACKGROUND: Dendritic cell immunotherapy has proven to be safe and induces an immune response in humans. We aimed to establish the efficacy of dendritic cells loaded with allogeneic tumour cell lysate (MesoPher, Amphera BV, 's-Hertogenbosch, Netherlands) as maintenance therapy in patients with pleural mesothelioma. METHODS: In this open-label, randomised, phase 2/3 study, patients with histologically confirmed unresectable pleural mesothelioma, aged 18 years or older, with an Eastern Cooperative Oncology Group performance status score of 0-1, and non-progressing disease after four to six cycles of standard chemotherapy (with pemetrexed 500 mg/m2 plus platinum [cisplatin 75 mg/m2 or carboplatin area under the curve of 5]) were recruited from four centres in Belgium, France, and The Netherlands. Participants were randomly assigned (1:1), using block randomisation (block size of 4), stratified by centre and histology (epithelioid vs other), to MesoPher treatment plus best supportive care or best supportive care alone. Patients received up to a maximum of five MesoPher infusions, with treatment administered on days 1, 15, and 29, and weeks 18 and 30. At each timepoint, participants received an injection of 25 × 106 dendritic cells (two-thirds of the dendritic cells were administered intravenously and a third were injected intradermally). Best supportive care was per local institutional standards. The primary endpoint was overall survival, assessed in all participants randomly assigned to treatment (full analysis set) and safety assessed in all randomly assigned participants, and who underwent leukapheresis if they were in the MesoPher group. This study is registered with ClinicalTrials.gov, NCT03610360, and is closed for accrual. FINDINGS: Between June 21, 2018, and June 10, 2021, 176 patients were screened and randomly assigned to the MesoPher group (n=88) or best supportive care alone group (n=88). One participant in the MesoPher group did not undergo leukapheresis. Mean age was 68 years (SD 8), 149 (85%) of 176 were male, 27 (15%) were female, 173 (98%) were White, two were Asian (1%), and one (1%) was other race. As of data cutoff (June 24, 2023), after a median follow up of 15·1 months (IQR 9·5-22·4), median overall survival was 16·8 months (95% CI 12·4-20·3; 61 [69%] of 88 died) in the MesoPher group and 18·3 months (14·3-21·9; 59 [67%] of 88 died) in the best supportive care group (hazard ratio 1·10 [95% CI 0·77-1·57]; log-rank p=0·62). The most common grade 3-4 treatment-emergent adverse events were chest pain (three [3%] of 87 in the MesoPher group vs two [2%] of 88 in the best supportive care group), dyspnoea (none vs two [2%]), anaemia (two [2%] vs none), nausea (none vs two [2%]), and pneumonia (none vs two [2%]). No deaths due to treatment-emergent adverse events were recorded. Treatment-related adverse events consisted of infusion-related reactions (fever, chills, and fatigue), which occurred in 64 (74%) of 87 patients in the MesoPher group, and injection-site reactions (itch, erythema, and induration), which occurred in 73 (84%) patients, and all were grade 1-2 in severity. No deaths were determined to be treatment related. INTERPRETATION: MesoPher did not show improvement in overall survival in patients with pleural mesothelioma. Immune checkpoint therapy is now standard of care in pleural mesothelioma. Further randomised studies are needed of combinations of MesoPher and immune checkpoint therapy, which might increase efficacy without adding major toxicities. FUNDING: Amphera BV and EU HORIZON.


Subject(s)
Dendritic Cells , Pleural Neoplasms , Humans , Female , Male , Dendritic Cells/transplantation , Dendritic Cells/immunology , Aged , Middle Aged , Pleural Neoplasms/therapy , Pleural Neoplasms/pathology , Pleural Neoplasms/mortality , Pleural Neoplasms/drug therapy , Pleural Neoplasms/immunology , Mesothelioma/therapy , Mesothelioma/drug therapy , Mesothelioma/pathology , Mesothelioma/mortality , Mesothelioma/immunology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Mesothelioma, Malignant/therapy , Mesothelioma, Malignant/pathology , Mesothelioma, Malignant/drug therapy , Maintenance Chemotherapy , Cisplatin/administration & dosage , Carboplatin/administration & dosage , Pemetrexed/administration & dosage
8.
J Clin Lab Anal ; 38(10): e25076, 2024 May.
Article in English | MEDLINE | ID: mdl-38853390

ABSTRACT

BACKGROUND: Severe acute pancreatitis (SAP) is associated with tremendous systemic inflammation, T-helper 17 (Th17) cells, and regulatory T (Treg) cells play an essential role in the inflammatory responses. Meanwhile, soluble fibrinogen-like protein 2 (Sfgl2) is a critical immunosuppressive effector cytokine of Treg cells and modulates immune responses. However, the impact of SAP induction on Sfgl2 expression and the role of Sfgl2 in immunomodulation under SAP conditions are largely unknown. METHODS: A taurocholate-induced mouse SAP model was established. The ratios of CD4+CD25+Foxp3+ Treg cells or CD4+IL-17+ Th17 cells in blood and pancreatic tissues as well as surface expression of CD80, CD86, and major histocompatibility complex class II (MHC-II) were determined by flow cytometry. Gene mRNA expression was determined by qPCR. Serum amylase and soluble factors were quantitated by commercial kits. Bone marrow-derived dendritic cells (DCs) were generated, and NF-κB/p65 translocation was measured by immunofluorescence staining. RESULTS: SAP induction in mice decreased the Th17/Treg ratio in the pancreatic tissue and increased the Th17/Treg ratio in the peripheral blood. In addition, SAP was associated with a reduced level of Sfgl2 in the pancreatic tissue and blood: higher levels of serum IL-17, IL-2, IFN-α, and TNF-α, and lower levels of serum IL-4 and IL-10. Furthermore, the SAP-induced reduction in Sfgl2 expression was accompanied by dysregulated maturation of bone marrow-derived DCs. CONCLUSIONS: SAP causes reduced Sfgl2 expression and Th17/Treg imbalance, thus providing critical insights for the development of Sfgl2- and Th17/Treg balance-targeted immunotherapies for patients with SAP.


Subject(s)
Disease Models, Animal , Fibrinogen , Pancreatitis , T-Lymphocytes, Regulatory , Taurocholic Acid , Th17 Cells , Animals , Th17 Cells/immunology , T-Lymphocytes, Regulatory/immunology , Pancreatitis/immunology , Pancreatitis/chemically induced , Pancreatitis/metabolism , Mice , Fibrinogen/metabolism , Male , Mice, Inbred C57BL , Down-Regulation , Dendritic Cells/immunology , Dendritic Cells/metabolism , Acute Disease , Pancreas/immunology , Pancreas/pathology , Pancreas/metabolism
9.
PLoS Genet ; 20(6): e1011313, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38870230

ABSTRACT

A quarter of humanity is estimated to have been exposed to Mycobacterium tuberculosis (Mtb) with a 5-10% risk of developing tuberculosis (TB) disease. Variability in responses to Mtb infection could be due to host or pathogen heterogeneity. Here, we focused on host genetic variation in a Peruvian population and its associations with gene regulation in monocyte-derived macrophages and dendritic cells (DCs). We recruited former household contacts of TB patients who previously progressed to TB (cases, n = 63) or did not progress to TB (controls, n = 63). Transcriptomic profiling of monocyte-derived DCs and macrophages measured the impact of genetic variants on gene expression by identifying expression quantitative trait loci (eQTL). We identified 330 and 257 eQTL genes in DCs and macrophages (False Discovery Rate (FDR) < 0.05), respectively. Four genes in DCs showed interaction between eQTL variants and TB progression status. The top eQTL interaction for a protein-coding gene was with FAH, the gene encoding fumarylacetoacetate hydrolase, which mediates the last step in mammalian tyrosine catabolism. FAH expression was associated with genetic regulatory variation in cases but not controls. Using public transcriptomic and epigenomic data of Mtb-infected monocyte-derived dendritic cells, we found that Mtb infection results in FAH downregulation and DNA methylation changes in the locus. Overall, this study demonstrates effects of genetic variation on gene expression levels that are dependent on history of infectious disease and highlights a candidate pathogenic mechanism through pathogen-response genes. Furthermore, our results point to tyrosine metabolism and related candidate TB progression pathways for further investigation.


Subject(s)
Dendritic Cells , Macrophages , Mycobacterium tuberculosis , Quantitative Trait Loci , Tuberculosis , Humans , Peru , Tuberculosis/genetics , Tuberculosis/microbiology , Macrophages/metabolism , Macrophages/microbiology , Mycobacterium tuberculosis/pathogenicity , Mycobacterium tuberculosis/genetics , Female , Dendritic Cells/metabolism , Male , Adult , Genetic Predisposition to Disease , Genetic Variation , Gene Expression Regulation , Middle Aged , Polymorphism, Single Nucleotide , Gene Expression Profiling
10.
Cell Mol Life Sci ; 81(1): 262, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38878186

ABSTRACT

Through Smad3-dependent signalings, transforming growth factor-ß (TGF-ß) suppresses the development, maturation, cytokine productions and cytolytic functions of NK cells in cancer. Silencing Smad3 remarkably restores the cytotoxicity of NK-92 against cancer in TGF-ß-rich microenvironment, but its effects on the immunoregulatory functions of NK cells remain obscure. In this study, we identified Smad3 functioned as a transcriptional repressor for CSF2 (GM-CSF) in NK cells. Therefore, disrupting Smad3 largely mitigated TGF-ß-mediated suppression on GM-CSF production by NK cells. Furthermore, silencing GM-CSF in Smad3 knockout NK cells substantially impaired their anti-lung carcinoma effects. In-depth study demonstrated that NK-derived GM-CSF strengthened T cell immune responses by stimulating dendritic cell differentiation and M1 macrophage polarization. Meanwhile, NK-derived GM-CSF promoted the survival of neutrophils, which in turn facilitated the terminal maturation of NK cells, and subsequently boosted NK-cell mediated cytotoxicity against lung carcinoma. Thus, Smad3-silenced NK-92 (NK-92-S3KD) may serve as a promising immunoadjuvant therapy with clinical translational value given its robust cytotoxicity against malignant cells and immunostimulatory functions to reinforce the therapeutic effects of other immunotherapies.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor , Killer Cells, Natural , Lung Neoplasms , Smad3 Protein , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Smad3 Protein/metabolism , Smad3 Protein/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Animals , Humans , Mice , Mice, Inbred C57BL , Cell Line, Tumor , Dendritic Cells/immunology , Dendritic Cells/metabolism , Transforming Growth Factor beta/metabolism , Cell Differentiation , Macrophages/metabolism , Macrophages/immunology , Signal Transduction
11.
Front Immunol ; 15: 1395596, 2024.
Article in English | MEDLINE | ID: mdl-38919629

ABSTRACT

Vascular calcification (VC) is considered a common pathological process in various vascular diseases. Accumulating studies have confirmed that VC is involved in the inflammatory response in heart disease, and SPP1+ macrophages play an important role in this process. In VC, studies have focused on the physiological and pathological functions of macrophages, such as pro-inflammatory or anti-inflammatory cytokines and pro-fibrotic vesicles. Additionally, macrophages and activated lymphocytes highly express SPP1 in atherosclerotic plaques, which promote the formation of fatty streaks and plaque development, and SPP1 is also involved in the calcification process of atherosclerotic plaques that results in heart failure, but the crosstalk between SPP1-mediated immune cells and VC has not been adequately addressed. In this review, we summarize the regulatory effect of SPP1 on VC in T cells, macrophages, and dendritic cells in different organs' VC, which could be a potential therapeutic target for VC.


Subject(s)
Macrophages , Osteopontin , Vascular Calcification , Humans , Osteopontin/metabolism , Vascular Calcification/immunology , Vascular Calcification/metabolism , Vascular Calcification/pathology , Animals , Macrophages/immunology , Macrophages/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/pathology , Plaque, Atherosclerotic/metabolism
12.
HLA ; 103(6): e15541, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38923358

ABSTRACT

Complications due to HCMV infection or reactivation remain a challenging clinical problem in immunocompromised patients, mainly due to insufficient or absent T-cell functionality. Knowledge of viral targets is crucial to improve monitoring of high-risk patients and optimise antiviral T-cell therapy. To expand the epitope spectrum, genetically-engineered dendritic cells (DCs) and fibroblasts were designed to secrete soluble (s)HLA-A*11:01 and infected with an HCMV mutant lacking immune evasion molecules (US2-6 + 11). More than 700 HLA-A*11:01-restricted epitopes, including more than 50 epitopes derived from a broad range of HCMV open-reading-frames (ORFs) were identified by mass spectrometry and screened for HLA-A*11:01-binding using established prediction tools. The immunogenicity of the 24 highest scoring new candidates was evaluated in vitro in healthy HLA-A*11:01+/HCMV+ donors. Thus, four subdominant epitopes and one immunodominant epitope, derived from the anti-apoptotic protein UL36 and ORFL101C (A11SAL), were identified. Their HLA-A*11:01 complex stability was verified in vitro. In depth analyses revealed highly proliferative and cytotoxic memory T-cell responses against A11SAL, with T-cell responses comparable to the immunodominant HLA-A*02:01-restricted HCMVpp65NLV epitope. A11SAL-specific T cells were also detectable in vivo in immunosuppressed transplant patients and shown to be effective in an in vitro HCMV-infection model, suggesting their crucial role in inhibiting viral replication and improvement of patient's outcome. The developed in vitro pipeline is the first to utilise genetically-engineered DCs to identify naturally presented immunodominant HCMV-derived epitopes. It therefore offers advantages over in silico predictions, is transferable to other HLA alleles, and will significantly expand the repertoire of viral targets to improve therapeutic options.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Dendritic Cells , Epitopes, T-Lymphocyte , Immunodominant Epitopes , Humans , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Immunodominant Epitopes/immunology , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/immunology , HLA-A11 Antigen/immunology , HLA-A11 Antigen/genetics , Fibroblasts/immunology , Fibroblasts/virology , Antigen-Presenting Cells/immunology
13.
Traffic ; 25(6): e12950, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38923715

ABSTRACT

Processes such as cell migration, phagocytosis, endocytosis, and exocytosis refer to the intense exchange of information between the internal and external environment in the cells, known as vesicular trafficking. In eukaryotic cells, these essential cellular crosstalks are controlled by Rab GTPases proteins through diverse adaptor proteins like SNAREs complex, coat proteins, phospholipids, kinases, phosphatases, molecular motors, actin, or tubulin cytoskeleton, among others, all necessary for appropriate mobilization of vesicles and distribution of molecules. Considering these molecular events, Rab GTPases are critical components in specific biological processes of immune cells, and many reports refer primarily to macrophages; therefore, in this review, we address specific functions in immune cells, concretely in the mechanism by which the GTPase contributes in dendritic cells (DCs) and, T/B lymphocytes.


Subject(s)
T-Lymphocytes , rab GTP-Binding Proteins , Humans , rab GTP-Binding Proteins/metabolism , Animals , T-Lymphocytes/metabolism , T-Lymphocytes/immunology , Dendritic Cells/metabolism , Dendritic Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/immunology
14.
Virulence ; 15(1): 2368080, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38899573

ABSTRACT

Dendritic cells (DCs) present an ideal target for delivering immunogenic cargo due to their potent antigen-presenting capabilities. This targeting approach holds promise in vaccine development by enhancing the efficiency of antigen recognition and capture by DCs. To identify a high-affinity targeting peptide binding to rabbit DCs, rabbit monocyte-derived DCs (raMoDCs) were isolated and cultured, and a novel peptide, HS (HSLRHDYGYPGH), was identified using a phage-displayed peptide library. Alongside HS, two other DC-targeting peptides, KC1 and MY, previously validated in our laboratory, were employed to construct recombinant Lactgobacillus reuteri fusion-expressed rabbit hemorrhagic disease virus (RHDV) capsid protein VP60. These recombinant Lactobacillus strains were named HS-VP60/L. reuteri, KC1-VP60/L. reuteri, and MY-VP60/L. reuteri. The ability of these recombinant Lactobacillus to bind rabbit DCs was evaluated both in vivo and in vitro. Results demonstrated that the DC-targeting peptide KC1 significantly enhanced the capture efficiency of recombinant Lactobacillus by raMoDCs, promoted DC maturation, and increased cytokine secretion. Furthermore, oral administration of KC1-VP60/L. reuteri effectively induced SIgA and IgG production in rabbits, prolonged rabbit survival post-challenge, and reduced RHDV copies in organs. In summary, the DC-targeting peptide KC1 exhibited robust binding to raMoDCs, and recombinant Lactobacillus expressing KC1-VP60 protein antigens efficiently induced systemic and mucosal immune responses in rabbits, conferring protective efficacy against RHDV. This study offers valuable insights for the development of novel RHDV vaccines.


Subject(s)
Dendritic Cells , Hemorrhagic Disease Virus, Rabbit , Limosilactobacillus reuteri , Peptides , Animals , Dendritic Cells/immunology , Rabbits , Hemorrhagic Disease Virus, Rabbit/immunology , Hemorrhagic Disease Virus, Rabbit/genetics , Limosilactobacillus reuteri/genetics , Limosilactobacillus reuteri/immunology , Peptides/immunology , Peptides/genetics , Caliciviridae Infections/prevention & control , Caliciviridae Infections/immunology , Reoviridae Infections/prevention & control , Reoviridae Infections/immunology , Capsid Proteins/genetics , Capsid Proteins/immunology , Viral Vaccines/immunology , Viral Vaccines/genetics , Lactobacillus/genetics , Lactobacillus/immunology
15.
Oncoimmunology ; 13(1): 2369373, 2024.
Article in English | MEDLINE | ID: mdl-38915784

ABSTRACT

Dendritic cells (DCs) are the main antigen presenting cells of the immune system and are essential for anti-tumor responses. DC-based immunotherapies are used in cancer treatment, but their functionality is not optimized and their clinical efficacy is currently limited. Approaches to improve DC functionality in anti-tumor immunity are therefore required. We have previously shown that the loss of ß2-integrin-mediated adhesion leads to epigenetic reprogramming of bone marrow-derived DCs (BM-DCs), resulting in an increased expression of costimulatory markers (CD86, CD80, and CD40), cytokines (IL-12) and the chemokine receptor CCR7. We now show that the loss of ß2-integrin-mediated adhesion of BM-DCs also leads to a generally suppressed metabolic profile, with reduced metabolic rate, decreased ROS production, and lowered glucose uptake in cells. The mRNA levels of glycolytic enzymes and glucose transporters were reduced, indicating transcriptional regulation of the metabolic phenotype. Surprisingly, although signaling through a central regulator of immune cell metabolisms, the mechanistic target of rapamycin (mTOR), was increased in BM-DCs with dysfunctional integrins, rapamycin treatment revealed that mTOR signaling was not involved in suppressing DC metabolism. Instead, bioinformatics and functional analyses showed that the Ikaros transcription factor may be involved in regulating the metabolic profile of non-adhesive DCs. Inversely, we found that induction of metabolic stress through treatment of cells with low levels of an inhibitor of glycolysis, 2-deoxyglucose (2DG), led to increased BM-DC activation. Specifically, 2DG treatment led to increased levels of Il-12 and Ccr7 mRNA, increased production of IL-12, increased levels of cell surface CCR7 and increased in vitro migration and T cell activation potential. Furthermore, 2DG treatment led to increased histone methylation in cells (H3K4me3, H3K27me3), indicating metabolic reprogramming. Finally, metabolic stress induced by 2DG treatment led to improved BM-DC-mediated anti-tumor responses in vivo in a melanoma cancer model, B16-OVA. In conclusion, our results indicate a role for ß2-integrin-mediated adhesion in regulating a novel type of metabolic reprogramming of DCs and DC-mediated anti-tumor responses, which may be targeted to enhance DC-mediated anti-tumor responses in cancer immunotherapy.


Subject(s)
CD18 Antigens , Dendritic Cells , Dendritic Cells/metabolism , Dendritic Cells/immunology , Animals , Mice , CD18 Antigens/metabolism , CD18 Antigens/genetics , Mice, Inbred C57BL , Cell Adhesion , Receptors, CCR7/metabolism , Receptors, CCR7/genetics , Melanoma, Experimental/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/genetics , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Humans , Metabolic Reprogramming
16.
Biomolecules ; 14(6)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38927060

ABSTRACT

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.


Subject(s)
5'-Nucleotidase , Adenosine , Dendritic Cells , Gingiva , Interferon-gamma , Mesenchymal Stem Cells , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Adenosine/metabolism , Interferon-gamma/metabolism , Gingiva/cytology , 5'-Nucleotidase/metabolism , Cells, Cultured , Apyrase/metabolism , GPI-Linked Proteins
17.
Front Immunol ; 15: 1393451, 2024.
Article in English | MEDLINE | ID: mdl-38903502

ABSTRACT

Dendritic cells (DCs) play a central role in the orchestration of effective T cell responses against tumors. However, their functional behavior is context-dependent. DC type, transcriptional program, location, intratumoral factors, and inflammatory milieu all impact DCs with regard to promoting or inhibiting tumor immunity. The following review introduces important facets of DC function, and how subset and phenotype can affect the interplay of DCs with other factors in the tumor microenvironment. It will also discuss how current cancer treatment relies on DC function, and survey the myriad ways with which immune therapy can more directly harness DCs to enact antitumor cytotoxicity.


Subject(s)
Dendritic Cells , Immunotherapy , Neoplasms , Tumor Microenvironment , Humans , Dendritic Cells/immunology , Neoplasms/therapy , Neoplasms/immunology , Tumor Microenvironment/immunology , Immunotherapy/methods , Animals
18.
J Vis Exp ; (207)2024 May 24.
Article in English | MEDLINE | ID: mdl-38856194

ABSTRACT

An advanced intestine-on-chip model recreating epithelial 3D organotypic villus-like and crypt-like structures has been developed. The immunocompetent model includes Human Umbilical Vein Endothelial Cells (HUVEC), Caco-2 intestinal epithelial cells, tissue-resident macrophages, and dendritic cells, which self-organize within the tissue, mirroring characteristics of the human intestinal mucosa. A unique aspect of this platform is its capacity to integrate circulating human primary immune cells, enhancing physiological relevance. The model is designed to investigate the intestinal immune system's response to bacterial and fungal colonization and infection. Due to its enlarged cavity size, the model offers diverse functional readouts such as permeation assays, cytokine release, and immune cell infiltration, and is compatible with immunofluorescence measurement of 3D structures formed by the epithelial cell layer. It hereby provides comprehensive insights into cell differentiation and function. The intestine-on-chip platform has demonstrated its potential in elucidating complex interactions between surrogates of a living microbiota and human host tissue within a microphysiological perfused biochip platform.


Subject(s)
Intestinal Mucosa , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/cytology , Caco-2 Cells , Human Umbilical Vein Endothelial Cells , Immunity, Mucosal/immunology , Lab-On-A-Chip Devices , Dendritic Cells/immunology , Dendritic Cells/cytology , Macrophages/immunology , Macrophages/cytology
19.
Arch Dermatol Res ; 316(7): 348, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849562

ABSTRACT

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


Subject(s)
Cytokines , Dendritic Cells , Glucosides , Monoterpenes , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Glucosides/pharmacology , Glucosides/therapeutic use , Animals , Cytokines/metabolism , Monoterpenes/pharmacology , Monoterpenes/therapeutic use , Humans , Mice , Dermatitis/drug therapy , Dermatitis/immunology , Dermatitis/metabolism , Interleukin-6/metabolism , Molecular Docking Simulation , Skin/pathology , Skin/drug effects , Skin/immunology , Skin/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Disease Models, Animal , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , Male , Thymic Stromal Lymphopoietin , Lymphocyte Activation/drug effects
20.
J Extracell Vesicles ; 13(6): e12446, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38844736

ABSTRACT

Dendritic cells (DCs) are essential orchestrators of immune responses and represent potential targets for immunomodulation in autoimmune diseases. Human amniotic fluid secretome is abundant in immunoregulatory factors, with extracellular vesicles (EVs) being a significant component. However, the impact of these EVs on dendritic cells subsets remain unexplored. In this study, we investigated the interaction between highly purified dendritic cell subsets and EVs derived from amniotic fluid stem cell lines (HAFSC-EVs). Our results suggest that HAFSC-EVs are preferentially taken up by conventional dendritic cell type 2 (cDC2) through CD29 receptor-mediated internalization, resulting in a tolerogenic DC phenotype characterized by reduced expression and production of pro-inflammatory mediators. Furthermore, treatment of cDC2 cells with HAFSC-EVs in coculture systems resulted in a higher proportion of T cells expressing the regulatory T cell marker Foxp3 compared to vehicle-treated control cells. Moreover, transfer of HAFSC-EV-treated cDC2s into an EAE mouse model resulted in the suppression of autoimmune responses and clinical improvement. These results suggest that HAFSC-EVs may serve as a promising tool for reprogramming inflammatory cDC2s towards a tolerogenic phenotype and for controlling autoimmune responses in the central nervous system, representing a potential platform for the study of the effects of EVs in DC subsets.


Subject(s)
Amniotic Fluid , Dendritic Cells , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental , Extracellular Vesicles , Multiple Sclerosis , Animals , Extracellular Vesicles/metabolism , Extracellular Vesicles/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Mice , Amniotic Fluid/cytology , Amniotic Fluid/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Humans , Multiple Sclerosis/therapy , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Female , Stem Cells/metabolism , Stem Cells/cytology , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...