Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 33.909
1.
Nat Commun ; 15(1): 3882, 2024 May 08.
Article En | MEDLINE | ID: mdl-38719809

In this randomized phase II clinical trial, we evaluated the effectiveness of adding the TLR agonists, poly-ICLC or resiquimod, to autologous tumor lysate-pulsed dendritic cell (ATL-DC) vaccination in patients with newly-diagnosed or recurrent WHO Grade III-IV malignant gliomas. The primary endpoints were to assess the most effective combination of vaccine and adjuvant in order to enhance the immune potency, along with safety. The combination of ATL-DC vaccination and TLR agonist was safe and found to enhance systemic immune responses, as indicated by increased interferon gene expression and changes in immune cell activation. Specifically, PD-1 expression increases on CD4+ T-cells, while CD38 and CD39 expression are reduced on CD8+ T cells, alongside an increase in monocytes. Poly-ICLC treatment amplifies the induction of interferon-induced genes in monocytes and T lymphocytes. Patients that exhibit higher interferon response gene expression demonstrate prolonged survival and delayed disease progression. These findings suggest that combining ATL-DC with poly-ICLC can induce a polarized interferon response in circulating monocytes and CD8+ T cells, which may represent an important blood biomarker for immunotherapy in this patient population.Trial Registration: ClinicalTrials.gov Identifier: NCT01204684.


CD8-Positive T-Lymphocytes , Cancer Vaccines , Carboxymethylcellulose Sodium/analogs & derivatives , Dendritic Cells , Glioma , Interferons , Poly I-C , Polylysine/analogs & derivatives , Humans , Dendritic Cells/immunology , Dendritic Cells/drug effects , Glioma/immunology , Glioma/therapy , Female , Male , Middle Aged , Cancer Vaccines/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/therapeutic use , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Poly I-C/administration & dosage , Poly I-C/pharmacology , Adult , Toll-Like Receptors/agonists , Imidazoles/pharmacology , Imidazoles/therapeutic use , Aged , Vaccination , Monocytes/immunology , Monocytes/drug effects , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Brain Neoplasms/drug therapy , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/drug effects , Immunotherapy/methods , Toll-Like Receptor Agonists
2.
Sci Immunol ; 9(95): eadi4191, 2024 May 10.
Article En | MEDLINE | ID: mdl-38728412

Conventional dendritic cells (DCs) are essential mediators of antitumor immunity. As a result, cancers have developed poorly understood mechanisms to render DCs dysfunctional within the tumor microenvironment (TME). After identification of CD63 as a specific surface marker, we demonstrate that mature regulatory DCs (mregDCs) migrate to tumor-draining lymph node tissues and suppress DC antigen cross-presentation in trans while promoting T helper 2 and regulatory T cell differentiation. Transcriptional and metabolic studies showed that mregDC functionality is dependent on the mevalonate biosynthetic pathway and its master transcription factor, SREBP2. We found that melanoma-derived lactate activates SREBP2 in tumor DCs and drives conventional DC transformation into mregDCs via homeostatic or tolerogenic maturation. DC-specific genetic silencing and pharmacologic inhibition of SREBP2 promoted antitumor CD8+ T cell activation and suppressed melanoma progression. CD63+ mregDCs were found to reside within the lymph nodes of several preclinical tumor models and in the sentinel lymph nodes of patients with melanoma. Collectively, this work suggests that a tumor lactate-stimulated SREBP2-dependent program promotes CD63+ mregDC development and function while serving as a promising therapeutic target for overcoming immune tolerance in the TME.


Dendritic Cells , Lactic Acid , Mice, Inbred C57BL , Signal Transduction , Sterol Regulatory Element Binding Protein 2 , Dendritic Cells/immunology , Animals , Mice , Humans , Sterol Regulatory Element Binding Protein 2/immunology , Lactic Acid/metabolism , Signal Transduction/immunology , Melanoma/immunology , Melanoma/pathology , Disease Progression , Immune Tolerance/immunology , Female , Cell Line, Tumor , Tumor Microenvironment/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology
3.
Front Immunol ; 15: 1372927, 2024.
Article En | MEDLINE | ID: mdl-38742105

The parasitic helminth Schistosoma mansoni is a potent inducer of type 2 immune responses by stimulating dendritic cells (DCs) to prime T helper 2 (Th2) responses. We previously found that S. mansoni soluble egg antigens (SEA) promote the synthesis of Prostaglandin E2 (PGE2) by DCs through ERK-dependent signaling via Dectin-1 and Dectin-2 that subsequently induces OX40L expression, licensing them for Th2 priming, yet the ligands present in SEA involved in driving this response and whether specific targeting of PGE2 synthesis by DCs could affect Th2 polarization are unknown. We here show that the ability of SEA to bind Dectin-2 and drive ERK phosphorylation, PGE2 synthesis, OX40L expression, and Th2 polarization is impaired upon cleavage of high-mannose glycans by Endoglycosidase H treatment. This identifies high-mannose glycans present on glycoproteins in SEA as important drivers of this signaling axis. Moreover, we find that OX40L expression and Th2 induction are abrogated when microsomal prostaglandin E synthase-1 (mPGES) is selectively inhibited, but not when a general COX-1/2 inhibitor is used. This shows that the de novo synthesis of PGE2 is vital for the Th2 priming function of SEA-stimulated DCs as well as points to the potential existence of other COX-dependent lipid mediators that antagonize PGE2-driven Th2 polarization. Lastly, specific PGE2 inhibition following immunization with S. mansoni eggs dampened the egg-specific Th cell response. In summary, our findings provide new insights in the molecular mechanisms underpinning Th2 induction by S. mansoni and identify druggable targets for potential control of helminth driven-Th2 responses.


Antigens, Helminth , Dendritic Cells , Dinoprostone , Lectins, C-Type , Mannose , Polysaccharides , Schistosoma mansoni , Th2 Cells , Animals , Schistosoma mansoni/immunology , Dinoprostone/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Lectins, C-Type/metabolism , Lectins, C-Type/immunology , Mannose/metabolism , Mannose/immunology , Mice , Polysaccharides/immunology , Polysaccharides/metabolism , Antigens, Helminth/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/metabolism , Schistosomiasis mansoni/parasitology , Ovum/immunology , Ovum/metabolism , Mice, Inbred C57BL , OX40 Ligand/metabolism
4.
Front Immunol ; 15: 1353922, 2024.
Article En | MEDLINE | ID: mdl-38745645

Introduction: During an innate inflammation, immune cells form distinct pro- and anti-inflammatory regions around pathogen-containing core-regions. Mast cells are localized in an anti-inflammatory microenvironment during the resolution of an innate inflammation, suggesting antiinflammatory roles of these cells. Methods: High-content imaging was used to investigated mast cell-dependent changes in the regional distribution of immune cells during an inflammation, induced by the toll-like receptor (TLR)-2 agonist zymosan. Results: The distance between the zymosan-containing core-region and the anti-inflammatory region, described by M2-like macrophages, increased in mast cell-deficient mice. Absence of mast cells abolished dendritic cell (DC) activation, as determined by CD86-expression and localized the DCs in greater distance to zymosan particles. The CD86- DCs had a higher expression of the pro-inflammatory interleukins (IL)-1ß and IL-12/23p40 as compared to activated CD86+ DCs. IL-4 administration restored CD86 expression, cytokine expression profile and localization of the DCs in mast cell-deficient mice. The IL-4 effects were mast cell-specific, since IL-4 reduction by eosinophil depletion did not affect activation of DCs. Discussion: We found that mast cells induce DC activation selectively at the site of inflammation and thereby determine their localization within the inflammation. Overall, mast cells have antiinflammatory functions in this inflammation model and limit the size of the pro-inflammatory region surrounding the zymosan-containing core region.


Dendritic Cells , Inflammation , Interleukin-4 , Mast Cells , Mice, Inbred C57BL , Toll-Like Receptor 2 , Zymosan , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Mice , Inflammation/immunology , Inflammation/metabolism , Interleukin-4/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/genetics , Mice, Knockout
5.
Front Immunol ; 15: 1374425, 2024.
Article En | MEDLINE | ID: mdl-38745644

Various gut bacteria, including Lactobacillus plantarum, possess several enzymes that produce hydroxy fatty acids (FAs), oxo FAs, conjugated FAs, and partially saturated FAs from polyunsaturated FAs as secondary metabolites. Among these derivatives, we identified 10-oxo-cis-6,trans-11-octadecadienoic acid (γKetoC), a γ-linolenic acid (GLA)-derived enon FA, as the most effective immunomodulator, which inhibited the antigen-induced immunoactivation and LPS-induced production of inflammatory cytokines. The treatment with γKetoC significantly suppressed proliferation of CD4+ T cells, LPS-induced activation of bone marrow-derived dendritic cells (BMDCs), and LPS-induced IL-6 release from peritoneal cells, splenocytes, and CD11c+ cells isolated from the spleen. γKetoC also inhibited the release of inflammatory cytokines from BMDCs stimulated with poly-I:C, R-848, or CpG. Further in vitro experiments using an agonist of GPR40/120 suggested the involvement of these GPCRs in the effects of γKetoC on DCs. We also found that γKetoC stimulated the NRF2 pathway in DCs, and the suppressive effects of γKetoC and agonist of GPR40/120 on the release of IL-6 and IL-12 were reduced in Nrf2-/- BMDCs. We evaluated the role of NRF2 in the anti-inflammatory effects of γKetoC in a dextran sodium sulfate-induced colitis model. The oral administration of γKetoC significantly reduced body weight loss, improved stool scores, and attenuated atrophy of the colon, in wild-type C57BL/6 and Nrf2+/- mice with colitis. In contrast, the pathology of colitis was deteriorated in Nrf2-/- mice even with the administration of γKetoC. Collectively, the present results demonstrated the involvement of the NRF2 pathway and GPCRs in γKetoC-mediated anti-inflammatory responses.


Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Mice, Inbred C57BL , NF-E2-Related Factor 2 , Receptors, G-Protein-Coupled , Signal Transduction , Animals , NF-E2-Related Factor 2/metabolism , Mice , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Gastrointestinal Microbiome/drug effects , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/immunology , Mice, Knockout , Cytokines/metabolism , Disease Models, Animal , Dextran Sulfate , Oleic Acids/pharmacology , Lactobacillus plantarum , Colitis/metabolism , Colitis/chemically induced , Colitis/drug therapy , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/drug effects , Male
6.
Oncoimmunology ; 13(1): 2349347, 2024.
Article En | MEDLINE | ID: mdl-38746870

The innate lymphoid cell (ILC) family is composed of heterogeneous innate effector and helper immune cells that preferentially reside in tissues where they promote tissue homeostasis. In cancer, they have been implicated in driving both pro- and anti-tumor responses. This apparent dichotomy highlights the need to better understand differences in the ILC composition and phenotype within different tumor types that could drive seemingly opposite anti-tumor responses. Here, we characterized the frequency and phenotype of various ILC subsets in melanoma metastases and primary epithelial ovarian tumors. We observed high PD-1 expression on ILC subsets isolated from epithelial ovarian tumor samples, while ILC populations in melanoma samples express higher levels of LAG-3. In addition, we found that the frequency of cytotoxic ILCs and NKp46+ILC3 in tumors positively correlates with monocytic cells and conventional type 2 dendritic cells, revealing potentially new interconnected immune cell subsets in the tumor microenvironment. Consequently, these observations may have direct relevance to tumor microenvironment composition and how ILC subset may influence anti-tumor immunity.


Carcinoma, Ovarian Epithelial , Immunity, Innate , Lymphocytes, Tumor-Infiltrating , Melanoma , Ovarian Neoplasms , Humans , Female , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Melanoma/immunology , Melanoma/pathology , Carcinoma, Ovarian Epithelial/immunology , Carcinoma, Ovarian Epithelial/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Tumor Microenvironment/immunology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Neoplasms, Glandular and Epithelial/immunology , Neoplasms, Glandular and Epithelial/pathology , Programmed Cell Death 1 Receptor/metabolism , Natural Cytotoxicity Triggering Receptor 1/metabolism , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/metabolism , Lymphocyte Activation Gene 3 Protein , Antigens, CD/metabolism
7.
Nat Commun ; 15(1): 4051, 2024 May 14.
Article En | MEDLINE | ID: mdl-38744839

Intestinal homeostasis is maintained by the response of gut-associated lymphoid tissue to bacteria transported across the follicle associated epithelium into the subepithelial dome. The initial response to antigens and how bacteria are handled is incompletely understood. By iterative application of spatial transcriptomics and multiplexed single-cell technologies, we identify that the double negative 2 subset of B cells, previously associated with autoimmune diseases, is present in the subepithelial dome in health. We show that in this location double negative 2 B cells interact with dendritic cells co-expressing the lupus autoantigens DNASE1L3 and C1q and microbicides. We observe that in humans, but not in mice, dendritic cells expressing DNASE1L3 are associated with sampled bacteria but not DNA derived from apoptotic cells. We propose that fundamental features of autoimmune diseases are microbiota-associated, interacting components of normal intestinal immunity.


B-Lymphocytes , Dendritic Cells , Endodeoxyribonucleases , Gastrointestinal Microbiome , Animals , Humans , Mice , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Gastrointestinal Microbiome/immunology , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/genetics , Dendritic Cells/immunology , Dendritic Cells/metabolism , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Female , Mice, Inbred C57BL , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Male
8.
Nano Lett ; 24(19): 5894-5903, 2024 May 15.
Article En | MEDLINE | ID: mdl-38709593

The combination of radiotherapy (RT) and immunotherapy shows promise in improving the clinical treatment of solid tumors; however, it faces challenges of low response rates and systemic toxicity. Herein, an implantable alginate/collagen hydrogel encapsulating C-C motif ligand 21 (CCL21)-expressing dendritic cells (CCL21-DCs@gel) was developed to potentiate the systemic antitumor effects of RT. The hydrogel functioned as a suitable reservoir for in vivo culture and proliferation of CCL21-DCs, thereby enabling sustained CCL21 release. The local CCL21 gradient induced by CCL21-DCs@gel significantly enhanced the efficacy of RT in suppressing primary tumor growth and inhibiting distant metastasis across several mouse models. Furthermore, the combination of RT with CCL21-DCs@gel provided complete prophylactic protection to mice. Mechanistic investigations revealed that CCL21-DCs@gel potentiated RT by promoting tumor lymphangiogenesis and attracting immune cell infiltration into the tumor. Collectively, these results suggest that CCL21-DCs@gel is a promising adjunct to RT for effectively eradicating tumors and preventing tumor recurrence.


Chemokine CCL21 , Dendritic Cells , Hydrogels , Animals , Hydrogels/chemistry , Mice , Dendritic Cells/drug effects , Dendritic Cells/immunology , Cell Line, Tumor , Humans , Alginates/chemistry , Neoplasms/radiotherapy , Neoplasms/pathology , Neoplasms/immunology , Collagen/chemistry , Immunotherapy/methods
9.
Ups J Med Sci ; 1292024.
Article En | MEDLINE | ID: mdl-38716077

Dendritic cells (DCs) possess a specialized function in presenting antigens and play pivotal roles in both innate and adaptive immune responses. Their ability to cross-present antigens from tumor cells to naïve T cells is instrumental in generating specific T-cell-mediated antitumor responses, crucial for controlling tumor growth and preventing tumor cell dissemination. However, within a tumor immune microenvironment (TIME), the functions of DCs can be significantly compromised. This review focuses on the profile, function, and activation of DCs, leveraging recent studies that reveal insights into their phenotype acquisition, transcriptional state, and functional programs through single-cell RNA sequence (scRNA-seq) analysis. Additionally, the therapeutic potential of DC-mediated tumor antigen sensing in priming antitumor immunity is discussed.


Dendritic Cells , Immunotherapy , Neoplasms , Tumor Microenvironment , Dendritic Cells/immunology , Humans , Neoplasms/immunology , Neoplasms/therapy , Immunotherapy/methods , Tumor Microenvironment/immunology , Antigens, Neoplasm/immunology , Animals
10.
Cell Commun Signal ; 22(1): 262, 2024 May 07.
Article En | MEDLINE | ID: mdl-38715122

Gene editing of living cells has become a crucial tool in medical research, enabling scientists to address fundamental biological questions and develop novel strategies for disease treatment. This technology has particularly revolutionized adoptive transfer cell therapy products, leading to significant advancements in tumor treatment and offering promising outcomes in managing transplant rejection, autoimmune disorders, and inflammatory diseases. While recent clinical trials have demonstrated the safety of tolerogenic dendritic cell (TolDC) immunotherapy, concerns remain regarding its effectiveness. This review aims to discuss the application of gene editing techniques to enhance the tolerance function of dendritic cells (DCs), with a particular focus on preclinical strategies that are currently being investigated to optimize the tolerogenic phenotype and function of DCs. We explore potential approaches for in vitro generation of TolDCs and provide an overview of emerging strategies for modifying DCs. Additionally, we highlight the primary challenges hindering the clinical adoption of TolDC therapeutics and propose future research directions in this field.


Autoimmune Diseases , Dendritic Cells , Humans , Dendritic Cells/immunology , Autoimmune Diseases/therapy , Autoimmune Diseases/immunology , Autoimmune Diseases/genetics , Animals , Gene Editing/methods , Immunotherapy/methods
11.
Sci Adv ; 10(19): eadm9561, 2024 May 10.
Article En | MEDLINE | ID: mdl-38718119

Lactic acid (LA) accumulation in the tumor microenvironment poses notable challenges to effective tumor immunotherapy. Here, an intelligent tumor treatment microrobot based on the unique physiological structure and metabolic characteristics of Veillonella atypica (VA) is proposed by loading Staphylococcus aureus cell membrane-coating BaTiO3 nanocubes (SAM@BTO) on the surface of VA cells (VA-SAM@BTO) via click chemical reaction. Following oral administration, VA-SAM@BTO accurately targeted orthotopic colorectal cancer through inflammatory targeting of SAM and hypoxic targeting of VA. Under in vitro ultrasonic stimulation, BTO catalyzed two reduction reactions (O2 → •O2- and CO2 → CO) and three oxidation reactions (H2O → •OH, GSH → GSSG, and LA → PA) simultaneously, effectively inducing immunogenic death of tumor cells. BTO catalyzed the oxidative coupling of VA cells metabolized LA, effectively disrupting the immunosuppressive microenvironment, improving dendritic cell maturation and macrophage M1 polarization, and increasing effector T cell proportions while decreasing regulatory T cell numbers, which facilitates synergetic catalysis and immunotherapy.


Colorectal Neoplasms , Immunotherapy , Tumor Microenvironment , Colorectal Neoplasms/therapy , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Immunotherapy/methods , Animals , Mice , Humans , Catalysis , Cell Line, Tumor , Nanostructures/chemistry , Biomimetic Materials/chemistry , Administration, Oral , Titanium/chemistry , Biomimetics/methods , Lactic Acid/chemistry , Dendritic Cells/immunology , Dendritic Cells/metabolism , Barium Compounds
12.
Balkan Med J ; 41(3): 174-185, 2024 May 03.
Article En | MEDLINE | ID: mdl-38700313

Background: Psoriasis is a chronic inflammatory skin disease that has no cure. While the specific cause of psoriasis is unknown, interactions between immune cells and inflammatory cytokines are believed to be important in its pathogenesis. Thymic stromal lymphopoietin (TSLP) is a cytokine produced by epithelial cells that profoundly affects dendritic cells (DCs) and is involved in allergy and inflammatory diseases. In some studies, its expression is higher in the skin of psoriasis patients, whereas it is increased in treated psoriasis patients when compared with untreated patients in others. Aims: To investigate the role of TSLP in the pathogenesis of psoriasis. Study Design: In vitro and in vivo study. Methods: To investigate the effect of TSLP on psoriasis in vivo, a mouse psoriasis model and shRNA targeting TSLP to reduce its expression were used. Mouse primary bone marrow dendritic cells (BMDCs) were cultured in vitro and used to investigate the signaling pathways activated by TSLP. Results: We found that reducing TSLP expression in psoriasis skin alleviated disease severity. TSLP activated the Janus kinase (JAK)/SYK pathway in psoriatic skin. In vitro studies with BMDCs demonstrated that TSLP increased DC maturation through the JAK/SYK pathway and activated DCs-secreted cytokines that stimulated CD4+ T cells to develop into T helper 17 (Th17) cells by activating STAT3 signaling. The JAK/SYK pathway inhibitor reduced the effect of TSLP on activating BMDCs and promoting Th17 differentiation by CD4+ T cells. Conclusion: These findings indicated that TSLP exerted its immune-modulating effect in psoriasis through the JAK/SYK pathway.


Cytokines , Dendritic Cells , Psoriasis , Th17 Cells , Thymic Stromal Lymphopoietin , Animals , Dendritic Cells/drug effects , Dendritic Cells/immunology , Mice , Cytokines/metabolism , Cytokines/drug effects , Th17 Cells/drug effects , Th17 Cells/immunology , Janus Kinases , Signal Transduction/drug effects , Disease Models, Animal , Syk Kinase , Humans
13.
Investig Clin Urol ; 65(3): 300-310, 2024 May.
Article En | MEDLINE | ID: mdl-38714521

PURPOSE: We developed immune checkpoint molecules to target recombinant dendritic cells (DCs) and verified their anti-tumor efficacy and immune response against prostate cancer. MATERIALS AND METHODS: DCs were generated from mononuclear cells in the tibia and femur bone marrow of mice. We knocked down the programmed death ligand 1 (PD-L1) on monocyte-derived DCs through siRNA PD-L1. Cell surface antigens were immune fluorescently stained through flow cytometry to analyze cultured cell phenotypes. Furthermore, we evaluated the efficacy of monocyte-derived DCs and recombinant DCs in a prostate cancer mouse model with subcutaneous TRAMP-C1 cells. Lastly, DC-induced mixed lymphocyte and lymphocyte-only proliferations were compared to determine cultured DCs' function. RESULTS: Compared to the control group, siRNA PD-L1 therapeutic DC-treated mice exhibited significantly inhibited tumor volume and increased tumor cell apoptosis. Remarkably, this treatment substantially augmented interferon-gamma and interleukin-2 production by stimulating T-cells in an allogeneic mixed lymphocyte reaction. Moreover, we demonstrated that PD-L1 gene silencing improved cell proliferation and cytokine production. CONCLUSIONS: We developed monocyte-derived DCs transfected with PD-L1 siRNA from mouse bone marrow. Our study highlights that PD-L1 inhibition in DCs increases antigen-specific immune responses, corroborating previous immunotherapy methodology findings regarding castration-resistant prostate cancer.


B7-H1 Antigen , Dendritic Cells , Prostatic Neoplasms , Dendritic Cells/immunology , Animals , Male , Mice , Prostatic Neoplasms/therapy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/genetics , Mice, Inbred C57BL , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods
14.
Sci Adv ; 10(18): eadn6537, 2024 May 03.
Article En | MEDLINE | ID: mdl-38701219

In mammals, males and females show marked differences in immune responses. Males are globally more sensitive to infectious diseases, while females are more susceptible to systemic autoimmunity. X-chromosome inactivation (XCI), the epigenetic mechanism ensuring the silencing of one X in females, may participate in these sex biases. We perturbed the expression of the trigger of XCI, the noncoding RNA Xist, in female mice. This resulted in reactivation of genes on the inactive X, including members of the Toll-like receptor 7 (TLR7) signaling pathway, in monocyte/macrophages and dendritic and B cells. Consequently, female mice spontaneously developed inflammatory signs typical of lupus, including anti-nucleic acid autoantibodies, increased frequencies of age-associated and germinal center B cells, and expansion of monocyte/macrophages and dendritic cells. Mechanistically, TLR7 signaling is dysregulated in macrophages, leading to sustained expression of target genes upon stimulation. These findings provide a direct link between maintenance of XCI and female-biased autoimmune manifestations and highlight altered XCI as a cause of autoimmunity.


Autoimmunity , Macrophages , Toll-Like Receptor 7 , X Chromosome Inactivation , Animals , Female , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , Autoimmunity/genetics , Mice , Male , Macrophages/metabolism , Macrophages/immunology , RNA, Long Noncoding/genetics , Signal Transduction , Dendritic Cells/immunology , Dendritic Cells/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology
15.
Front Immunol ; 15: 1360140, 2024.
Article En | MEDLINE | ID: mdl-38711513

Introduction: Modified Vaccinia Virus Ankara (MVA) is a safe vaccine vector inducing long- lasting and potent immune responses. MVA-mediated CD8+T cell responses are optimally induced, if both, direct- and cross-presentation of viral or recombinant antigens by dendritic cells are contributing. Methods: To improve the adaptive immune responses, we investigated the role of the purinergic receptor P2X7 (P2RX7) in MVA-infected feeder cells as a modulator of cross-presentation by non-infected dendritic cells. The infected feeder cells serve as source of antigen and provide signals that help to attract dendritic cells for antigen take up and to license these cells for cross-presentation. Results: We demonstrate that presence of an active P2RX7 in major histocompatibility complex (MHC) class I (MHCI) mismatched feeder cells significantly enhanced MVA-mediated antigen cross-presentation. This was partly regulated by P2RX7-specific processes, such as the increased availability of extracellular particles as well as the altered cellular energy metabolism by mitochondria in the feeder cells. Furthermore, functional P2RX7 in feeder cells resulted in a delayed but also prolonged antigen expression after infection. Discussion: We conclude that a combination of the above mentioned P2RX7-depending processes leads to significantly increased T cell activation via cross- presentation of MVA-derived antigens. To this day, P2RX7 has been mostly investigated in regards to neuroinflammatory diseases and cancer progression. However, we report for the first time the crucial role of P2RX7 for antigen- specific T cell immunity in a viral infection model.


CD8-Positive T-Lymphocytes , Cross-Priming , Dendritic Cells , Receptors, Purinergic P2X7 , Vaccinia virus , Receptors, Purinergic P2X7/immunology , Receptors, Purinergic P2X7/metabolism , Cross-Priming/immunology , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Vaccinia virus/immunology , Mice , CD8-Positive T-Lymphocytes/immunology , Genetic Vectors , Mice, Inbred C57BL , Antigen Presentation/immunology , Antigens, Viral/immunology , Humans , Viral Vaccines/immunology
16.
Front Immunol ; 15: 1392316, 2024.
Article En | MEDLINE | ID: mdl-38711516

Streptococcus pneumoniae remains a significant global threat, with existing vaccines having important limitations such as restricted serotype coverage and high manufacturing costs. Pneumococcal lipoproteins are emerging as promising vaccine candidates due to their surface exposure and conservation across various serotypes. While prior studies have explored their potential in mice, data in a human context and insights into the impact of the lipid moiety remain limited. In the present study, we examined the immunogenicity of two pneumococcal lipoproteins, DacB and MetQ, both in lipidated and non-lipidated versions, by stimulation of primary human immune cells. Immune responses were assessed by the expression of common surface markers for activation and maturation as well as cytokines released into the supernatant. Our findings indicate that in the case of MetQ lipidation was crucial for activation of human antigen-presenting cells such as dendritic cells and macrophages, while non-lipidated DacB demonstrated an intrinsic potential to induce an innate immune response. Nevertheless, immune responses to both proteins were enhanced by lipidation. Interestingly, following stimulation of dendritic cells with DacB, LipDacB and LipMetQ, cytokine levels of IL-6 and IL-23 were significantly increased, which are implicated in triggering potentially important Th17 cell responses. Furthermore, LipDacB and LipMetQ were able to induce proliferation of CD4+ T cells indicating their potential to induce an adaptive immune response. These findings contribute valuable insights into the immunogenic properties of pneumococcal lipoproteins, emphasizing their potential role in vaccine development against pneumococcal infections.


Adaptive Immunity , Bacterial Proteins , Cytokines , Streptococcus pneumoniae , Humans , Streptococcus pneumoniae/immunology , Cytokines/metabolism , Bacterial Proteins/immunology , Lipoproteins/immunology , Lipoproteins/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Pneumococcal Vaccines/immunology , Pneumococcal Infections/immunology , Pneumococcal Infections/prevention & control , Macrophages/immunology , Macrophages/metabolism , Cells, Cultured
17.
ACS Nano ; 18(19): 12194-12209, 2024 May 14.
Article En | MEDLINE | ID: mdl-38689426

In situ vaccines (ISVs) utilize the localized delivery of chemotherapeutic agents or radiotherapy to stimulate the release of endogenous antigens from tumors, thereby eliciting systemic and persistent immune activation. Recently, a bioinspired ISV strategy has attracted tremendous attention due to its features such as an immune adjuvant effect and genetic plasticity. M13 bacteriophages are natural nanomaterials with intrinsic immunogenicity, genetic flexibility, and cost-effectiveness for large-scale production, demonstrating the potential for application in cancer vaccines. In this study, we propose an ISV based on the engineered M13 bacteriophage targeting CD40 (M13CD40) for dendritic cell (DC)-targeted immune stimulation, named H-GM-M13CD40. We induce immunogenic cell death and release tumor antigens through local delivery of (S)-10-hydroxycamptothecin (HCPT), followed by intratumoral injection of granulocyte-macrophage colony stimulating factor (GM-CSF) and M13CD40 to enhance DC recruitment and activation. We demonstrate that this ISV strategy can result in significant accumulation and activation of DCs at the tumor site, reversing the immunosuppressive tumor microenvironment. In addition, H-GM-M13CD40 can synergize with the PD-1 blockade and induce abscopal effects in cold tumor models. Overall, our study verifies the immunogenicity of the engineered M13CD40 bacteriophage and provides a proof of concept that the engineered M13CD40 phage can function as an adjuvant for ISVs.


Bacteriophage M13 , Cancer Vaccines , Dendritic Cells , Tumor Microenvironment , Cancer Vaccines/immunology , Tumor Microenvironment/immunology , Tumor Microenvironment/drug effects , Animals , Bacteriophage M13/immunology , Bacteriophage M13/chemistry , Mice , Dendritic Cells/immunology , CD40 Antigens/immunology , CD40 Antigens/metabolism , Mice, Inbred C57BL , Female , Cell Line, Tumor , Granulocyte-Macrophage Colony-Stimulating Factor , Antigens, Neoplasm/immunology , Humans
18.
ACS Nano ; 18(19): 12295-12310, 2024 May 14.
Article En | MEDLINE | ID: mdl-38695532

Immune checkpoint blockade (ICB) has brought tremendous clinical progress, but its therapeutic outcome can be limited due to insufficient activation of dendritic cells (DCs) and insufficient infiltration of cytotoxic T lymphocytes (CTLs). Evoking immunogenic cell death (ICD) is one promising strategy to promote DC maturation and elicit T-cell immunity, whereas low levels of ICD induction of solid tumors restrict durable antitumor efficacy. Herein, we report a genetically edited cell membrane-coated cascade nanozyme (gCM@MnAu) for enhanced cancer immunotherapy by inducing ICD and activating the stimulator of the interferon genes (STING) pathway. In the tumor microenvironment (TME), the gCM@MnAu initiates a cascade reaction and generates abundant cytotoxic hydroxyl (•OH), resulting in improved chemodynamic therapy (CDT) and boosted ICD activation. In addition, released Mn2+ during the cascade reaction activates the STING pathway and further promotes the DC maturation. More importantly, activated immunogenicity in the TME significantly improves gCM-mediated PD-1/PD-L1 checkpoint blockade therapy by eliciting systemic antitumor responses. In breast cancer subcutaneous and lung metastasis models, the gCM@MnAu showed synergistically enhanced therapeutic effects and significantly prolonged the survival of mice. This work develops a genetically edited nanozyme-based therapeutic strategy to improve DC-mediated cross-priming of T cells against poorly immunogenic solid tumors.


Immunotherapy , Animals , Mice , Female , Humans , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Mice, Inbred BALB C , Cell Line, Tumor , Immunogenic Cell Death/drug effects , Membrane Proteins/genetics , Membrane Proteins/immunology , Nanoparticles/chemistry
19.
Nat Commun ; 15(1): 3926, 2024 May 09.
Article En | MEDLINE | ID: mdl-38724513

Patients with decreased levels of CD18 (ß2 integrins) suffer from life-threatening bacterial and fungal infections. CD11b, the α subunit of integrin CR3 (CD11b/CD18, αMß2), is essential for mice to fight against systemic Candida albicans infections. Live elongating C. albicans activates CR3 in immune cells. However, the hyphal ligands that activate CR3 are not well defined. Here, we discovered that the C. albicans Als family proteins are recognized by the I domain of CD11b in macrophages. This recognition synergizes with the ß-glucan-bound lectin-like domain to activate CR3, thereby promoting Syk signaling and inflammasome activation. Dectin-2 activation serves as the "outside-in signaling" for CR3 activation at the entry site of incompletely sealed phagosomes, where a thick cuff of F-actin forms to strengthen the local interaction. In vitro, CD18 partially contributes to IL-1ß release from dendritic cells induced by purified hyphal Als3. In vivo, Als3 is vital for C. albicans clearance in mouse kidneys. These findings uncover a novel family of ligands for the CR3 I domain that promotes fungal clearance.


CD18 Antigens , Candidiasis , Fungal Proteins , Lectins, C-Type , Macrophages , Animals , Mice , beta-Glucans/metabolism , beta-Glucans/immunology , Candida albicans/immunology , Candidiasis/immunology , Candidiasis/microbiology , CD11b Antigen/metabolism , CD11b Antigen/immunology , CD18 Antigens/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Fungal Proteins/metabolism , Fungal Proteins/immunology , Lectins, C-Type/metabolism , Lectins, C-Type/immunology , Macrophages/immunology , Macrophages/metabolism , Signal Transduction
20.
Cell ; 187(10): 2521-2535.e21, 2024 May 09.
Article En | MEDLINE | ID: mdl-38697107

Cancer immunotherapy remains limited by poor antigenicity and a regulatory tumor microenvironment (TME). Here, we create "onion-like" multi-lamellar RNA lipid particle aggregates (LPAs) to substantially enhance the payload packaging and immunogenicity of tumor mRNA antigens. Unlike current mRNA vaccine designs that rely on payload packaging into nanoparticle cores for Toll-like receptor engagement in immune cells, systemically administered RNA-LPAs activate RIG-I in stromal cells, eliciting massive cytokine/chemokine response and dendritic cell/lymphocyte trafficking that provokes cancer immunogenicity and mediates rejection of both early- and late-stage murine tumor models. In client-owned canines with terminal gliomas, RNA-LPAs improved survivorship and reprogrammed the TME, which became "hot" within days of a single infusion. In a first-in-human trial, RNA-LPAs elicited rapid cytokine/chemokine release, immune activation/trafficking, tissue-confirmed pseudoprogression, and glioma-specific immune responses in glioblastoma patients. These data support RNA-LPAs as a new technology that simultaneously reprograms the TME while eliciting rapid and enduring cancer immunotherapy.


Immunotherapy , Tumor Microenvironment , Animals , Immunotherapy/methods , Mice , Dogs , Humans , Dendritic Cells/immunology , Dendritic Cells/metabolism , Cytokines/metabolism , Glioblastoma/therapy , Glioblastoma/immunology , Mice, Inbred C57BL , Female , Glioma/therapy , Glioma/immunology , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , RNA, Messenger/metabolism , RNA, Messenger/genetics , RNA/metabolism , RNA/therapeutic use , Cell Line, Tumor , Neoplasms/therapy , Neoplasms/immunology , Brain Neoplasms/therapy , Brain Neoplasms/immunology
...