Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Cancer Immunol Immunother ; 72(12): 4385-4397, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37938368

ABSTRACT

This study examined the composition of the immune microenvironment at different sites within resected pancreas specimens from patients with pancreatic ductal adenocarcinoma (PDAC). Therefore, single-cell suspensions were made from fresh tumor and non-tumorous tissue. Fourteen patients were included from whom twelve PDAC and five non-tumorous samples were obtained. These samples were analyzed with a nineteen marker panel on the Aurora spectral flow cytometer. Furthermore, slides from formalin-fixed paraffine PDACs of eight additional patients were stained with eight markers and analyzed by multispectral imaging. These corresponded to central tumor, periphery of the tumor, i.e., invasive front and resected lymph node and were divided into tumor and adjacent tissue. In the single-cell suspension, a decreased ratio between lymphoid and myeloid cells and between M1 and M2 macrophages was observed in the tumor tissue compared to non-tumorous tissue. Furthermore, an increase in CD169 + macrophages in patients undergoing neoadjuvant therapy was found. Using immunofluorescence, more macrophages compared to T cells were observed, as well as a lower ratio of CD8 to M2 macrophage, a higher ratio of CD4-CD8 T cells and a higher ratio of immune-suppressive cells to pro-inflammatory cells in the PDAC area compared to the adjacent non-tumorous tissue. Finally, there were more immune-suppressive cells in the central tumor area compared to the invasive front. In conclusion, we show a gradient in the immune-suppressive environment in PDAC from most suppressive in the central tumor to least suppressive in distant non-tumorous tissue.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Tumor Microenvironment , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Pancreas/pathology , T-Lymphocytes
2.
Proc Natl Acad Sci U S A ; 117(34): 20706-20716, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32764145

ABSTRACT

Cytotoxic T cell differentiation is guided by epigenome adaptations, but how epigenetic mechanisms control lymphocyte development has not been well defined. Here we show that the histone methyltransferase DOT1L, which marks the nucleosome core on active genes, safeguards normal differentiation of CD8+ T cells. T cell-specific ablation of Dot1L resulted in loss of naïve CD8+ T cells and premature differentiation toward a memory-like state, independent of antigen exposure and in a cell-intrinsic manner. Mechanistically, DOT1L controlled CD8+ T cell differentiation by ensuring normal T cell receptor density and signaling. DOT1L also maintained epigenetic identity, in part by indirectly supporting the repression of developmentally regulated genes. Finally, deletion of Dot1L in T cells resulted in an impaired immune response. Through our study, DOT1L is emerging as a central player in physiology of CD8+ T cells, acting as a barrier to prevent premature differentiation and controlling epigenetic integrity.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Cell Differentiation/genetics , Epigenesis, Genetic/genetics , Epigenomics , Female , Histone Methyltransferases/metabolism , Histone-Lysine N-Methyltransferase/physiology , Histones/metabolism , Male , Methyltransferases/metabolism , Mice
3.
Proc Natl Acad Sci U S A ; 117(44): 27528-27539, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33067394

ABSTRACT

Priming of CD8+ T cells by dendritic cells (DCs) is crucial for the generation of effective antitumor immune responses. Here, we describe a liposomal vaccine carrier that delivers tumor antigens to human CD169/Siglec-1+ antigen-presenting cells using gangliosides as targeting ligands. Ganglioside-liposomes specifically bound to CD169 and were internalized by in vitro-generated monocyte-derived DCs (moDCs) and macrophages and by ex vivo-isolated splenic macrophages in a CD169-dependent manner. In blood, high-dimensional reduction analysis revealed that ganglioside-liposomes specifically targeted CD14+ CD169+ monocytes and Axl+ CD169+ DCs. Liposomal codelivery of tumor antigen and Toll-like receptor ligand to CD169+ moDCs and Axl+ CD169+ DCs led to cytokine production and robust cross-presentation and activation of tumor antigen-specific CD8+ T cells. Finally, Axl+ CD169+ DCs were present in cancer patients and efficiently captured ganglioside-liposomes. Our findings demonstrate a nanovaccine platform targeting CD169+ DCs to drive antitumor T cell responses.


Subject(s)
Cancer Vaccines/administration & dosage , Dendritic Cells/immunology , Macrophages/immunology , Neoplasms/therapy , Vaccination/methods , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cross-Priming/immunology , Dendritic Cells/metabolism , Gangliosides , Humans , Immunogenicity, Vaccine , Leukocytes, Mononuclear , Liposomes , Macrophages/metabolism , Neoplasms/immunology , Primary Cell Culture , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Sialic Acid Binding Ig-like Lectin 1/metabolism , THP-1 Cells , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Axl Receptor Tyrosine Kinase
4.
FASEB J ; 34(9): 12406-12418, 2020 09.
Article in English | MEDLINE | ID: mdl-33411367

ABSTRACT

The clinically successful adjuvant MF59 is used in seasonal influenza vaccines, which is proposed to enhance immunity by creating an immune-competent microenvironment in the muscle that allows recruitment of immune cells that drive adaptive immune responses. Here, we examined whether the clinically successful adjuvants MF59/AddaVax could be used for subcutaneous use and how antigen delivery can be synergized with cellular dynamics at the vaccination site. Subcutaneous injection of AddaVax leads to thickening of the skin, characterized by a neutrophil-monocyte recruitment sequence. Skin-infiltrating CCR2+Ly6Chigh monocytes showed differentiation to CD11b+Ly6C+MHCII+CD11c+CD64+ monocyte-derived DCs over time in the hypodermal layers of the skin, expressing high levels of CD209a/mDC-SIGN. Surprisingly, skin thickening was accompanied with increased white adipose tissue highly enriched with monocytes. Analysis of the skin-draining lymph nodes revealed early increases in neutrophils and moDCs at 12 hours after injection and later increases in migratory cDC2s. Subcutaneous vaccination with AddaVax enhanced antigen-specific CD8+ and CD4+ T cell responses, while moDC targeting using antigen-coupled CD209a antibody additionally boosted humoral responses. Hence, oil-in-water emulsions provide an attractive immune modulatory adjuvants aimed at increasing cellular responses, as well as antibody responses when combined with moDC targeting.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Influenza Vaccines/immunology , Polysorbates/administration & dosage , Skin/immunology , Squalene/administration & dosage , Animals , Dendritic Cells/immunology , Influenza Vaccines/administration & dosage , Mice , Mice, Inbred C57BL , Monocytes/physiology , Neutrophils/physiology , T-Lymphocytes/immunology , Vaccination
5.
J Immunol ; 202(1): 171-182, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30504420

ABSTRACT

Innate lymphoid cells (ILCs) guard epithelial tissue integrity during homeostasis, but can be potent immune effector cells during inflammation. Precursors to all ILC subsets (ILC precursors [ILCP]) have been identified in human peripheral blood (PB). We found that during homeostasis, ILCP in PB of mouse and human expressed homing receptors for secondary lymphoid organs, mainly CD62L. These ILCP entered mouse lymph nodes in a CD62L-dependent way and relied on S1P receptors for their exit. Importantly, CD62L expression was absent on human ILCs expressing NKp44 in tonsils and PB of Crohn disease patients, and relatively fewer CD62L+ ILCP were present in PB of Crohn disease patients. These data are in agreement with selective expression of CD62L on nonactivated ILCP. As such, we conclude that CD62L not only serves as a functional marker of ILCP, but has potential to be used in the clinic as a diagnostic marker in inflammatory disorders.


Subject(s)
Blood Cells/immunology , Crohn Disease/immunology , L-Selectin/metabolism , Lymph Nodes/immunology , Lymphocytes/immunology , Lymphoid Progenitor Cells/physiology , Animals , Cells, Cultured , Female , Homeostasis , Humans , Immunity, Innate , L-Selectin/genetics , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Natural Cytotoxicity Triggering Receptor 2/metabolism , Receptors, Lysosphingolipid/metabolism
6.
PLoS Pathog ; 13(7): e1006539, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28753651

ABSTRACT

Infection with the helminth Schistosoma (S.) mansoni drives the development of interleukin (IL)-10-producing regulatory B (Breg) cells in mice and man, which have the capacity to reduce experimental allergic airway inflammation and are thus of high therapeutic interest. However, both the involved antigen and cellular mechanisms that drive Breg cell development remain to be elucidated. Therefore, we investigated whether S. mansoni soluble egg antigens (SEA) directly interact with B cells to enhance their regulatory potential, or act indirectly on B cells via SEA-modulated macrophage subsets. Intraperitoneal injections of S. mansoni eggs or SEA significantly upregulated IL-10 and CD86 expression by marginal zone B cells. Both B cells as well as macrophages of the splenic marginal zone efficiently bound SEA in vivo, but macrophages were dispensable for Breg cell induction as shown by macrophage depletion with clodronate liposomes. SEA was internalized into acidic cell compartments of B cells and induced a 3-fold increase of IL-10, which was dependent on endosomal acidification and was further enhanced by CD40 ligation. IPSE/alpha-1, one of the major antigens in SEA, was also capable of inducing IL-10 in naïve B cells, which was reproduced by tobacco plant-derived recombinant IPSE. Other major schistosomal antigens, omega-1 and kappa-5, had no effect. SEA depleted of IPSE/alpha-1 was still able to induce Breg cells indicating that SEA contains more Breg cell-inducing components. Importantly, SEA- and IPSE-induced Breg cells triggered regulatory T cell development in vitro. SEA and recombinant IPSE/alpha-1 also induced IL-10 production in human CD1d+ B cells. In conclusion, the mechanism of S. mansoni-induced Breg cell development involves a direct targeting of B cells by SEA components such as the secretory glycoprotein IPSE/alpha-1.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Egg Proteins/immunology , Helminth Proteins/immunology , Ovum/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Animals , Antigens, Helminth/genetics , Antigens, Helminth/immunology , Egg Proteins/genetics , Female , Helminth Proteins/genetics , Humans , Interleukin-10/immunology , Mice , Mice, Inbred C57BL , Schistosoma mansoni/genetics , Schistosomiasis mansoni/genetics , Schistosomiasis mansoni/parasitology
7.
Proc Natl Acad Sci U S A ; 113(12): 3329-34, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26941238

ABSTRACT

Sialic acids are negatively charged nine-carbon carboxylated monosaccharides that often cap glycans on glycosylated proteins and lipids. Because of their strategic location at the cell surface, sialic acids contribute to interactions that are critical for immune homeostasis via interactions with sialic acid-binding Ig-type lectins (siglecs). In particular, these interactions may be of importance in cases where sialic acids may be overexpressed, such as on certain pathogens and tumors. We now demonstrate that modification of antigens with sialic acids (Sia-antigens) regulates the generation of antigen-specific regulatory T (Treg) cells via dendritic cells (DCs). Additionally, DCs that take up Sia-antigen prevent formation of effector CD4(+) and CD8(+)T cells. Importantly, the regulatory properties endowed on DCs upon Sia-antigen uptake are antigen-specific: only T cells responsive to the sialylated antigen become tolerized. In vivo, injection of Sia-antigen-loaded DCs increased de novo Treg-cell numbers and dampened effector T-cell expansion and IFN-γ production. The dual tolerogenic features that Sia-antigen imposed on DCs are Siglec-E-mediated and maintained under inflammatory conditions. Moreover, loading DCs with Sia-antigens not only inhibited the function of in vitro-established Th1 and Th17 effector T cells but also significantly dampened ex vivo myelin-reactive T cells, present in the circulation of mice with experimental autoimmune encephalomyelitis. These data indicate that sialic acid-modified antigens instruct DCs in an antigen-specific tolerogenic programming, enhancing Treg cells and reducing the generation and propagation of inflammatory T cells. Our data suggest that sialylation of antigens provides an attractive way to induce antigen-specific immune tolerance.


Subject(s)
Adaptation, Physiological/immunology , Antigens/immunology , Cell Proliferation , N-Acetylneuraminic Acid/chemistry , T-Lymphocytes, Regulatory/immunology , Animals , Antigens/chemistry , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/cytology
8.
EMBO J ; 31(1): 201-13, 2012 Jan 04.
Article in English | MEDLINE | ID: mdl-22036947

ABSTRACT

Early during Gram-negative sepsis, excessive release of pro-inflammatory cytokines can cause septic shock that is often followed by a state of immune paralysis characterized by the failure to mount adaptive immunity towards secondary microbial infections. Especially, the early mechanisms responsible for such immune hypo-responsiveness are unclear. Here, we show that TLR4 is the key immune sensing receptor to initiate paralysis of T-cell immunity after bacterial sepsis. Downstream of TLR4, signalling through TRIF but not MyD88 impaired the development of specific T-cell immunity against secondary infections. We identified type I interferon (IFN) released from splenic macrophages as the critical factor causing T-cell immune paralysis. Early during sepsis, type I IFN acted selectively on dendritic cells (DCs) by impairing antigen presentation and secretion of pro-inflammatory cytokines. Our results reveal a novel immune regulatory role for type I IFN in the initiation of septic immune paralysis, which is distinct from its well-known immune stimulatory effects. Moreover, we identify potential molecular targets for therapeutic intervention to overcome impairment of T-cell immunity after sepsis.


Subject(s)
Adaptive Immunity , Interferon Type I/metabolism , Macrophages/metabolism , Sepsis/immunology , Spleen/metabolism , Animals , Dendritic Cells/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/metabolism , Sepsis/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism
9.
Eur J Immunol ; 45(3): 747-57, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25487358

ABSTRACT

Ag delivery to specific APCs is an attractive approach in developing strategies for vaccination. CD169(+) macrophages in the marginal zone of the spleen represent a suitable target for delivery of Ag because of their strategic location, which is optimal for the capture of blood-borne Ag and their close proximity to B cells and T cells in the white pulp. Here we show that Ag targeting to CD169(+) macrophages in mice resulted in strong, isotype-switched, high-affinity Ab production and the preferential induction and long-term persistence of Ag-specific GC B cells and follicular Th cells. In agreement with these observations, CD169(+) macrophages retained intact Ag, induced cognate activation of B cells, and increased expression of costimulatory molecules upon activation. In addition, macrophages were required for the production of cytokines that promote B-cell responses. Our results identify CD169(+) macrophages as promoters of high-affinity humoral immune responses and emphasize the value of CD169 as target for Ag delivery to improve vaccine responses.


Subject(s)
Antibody Formation/physiology , B-Lymphocytes/immunology , Germinal Center/immunology , Lymphocyte Activation/physiology , Sialic Acid Binding Ig-like Lectin 1/immunology , Spleen/immunology , Animals , B-Lymphocytes/cytology , Germinal Center/cytology , Mice , Mice, Mutant Strains , Sialic Acid Binding Ig-like Lectin 1/genetics , Spleen/cytology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology
10.
Eur J Immunol ; 45(1): 89-100, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25311225

ABSTRACT

The vitamin A metabolite retinoic acid (RA) has been reported to suppress Th1 responses and enhance Th2 responses. Here, we investigated whether differences in vitamin A metabolism could underlie the differences between C57BL/6 and BALB/c mice, which are reportedly seen as Th1 and Th2 responders, respectively. BALB/c mice were shown to have higher intestinal epithelial expression of RALDH1 (where RALDH is retinaldehyde dehydrogenase), and, consequently, higher RALDH activity in MLN-DCs, leading to an increased ability to induce IgA class switching in B cells. Furthermore, within BALB/c mice, induction of IgA secretion as well as increased accumulation of regulatory T cells (Treg) in the intestinal lamina propria was observed. Additionally, as BALB/c mice are more resistant to dextran sulphate sodium (DSS) induced colitis, mice that lacked vitamin A in their diet had a more severe form of DSS-induced colitis compared to control mice. Therefore, the level of RA production and consequently the degree of RA-mediated signaling is crucial for the efficiency of the mucosal immune system.


Subject(s)
Colitis/immunology , Immunity, Mucosal , Intestines/immunology , Isoenzymes/immunology , Mucous Membrane/immunology , Retinal Dehydrogenase/immunology , Vitamin A/metabolism , Aldehyde Dehydrogenase 1 Family , Animals , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Dextran Sulfate , Gene Expression , Immunoglobulin A/genetics , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Immunoglobulin Class Switching , Intestinal Mucosa/metabolism , Intestines/pathology , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mucous Membrane/metabolism , Mucous Membrane/pathology , Retinal Dehydrogenase/genetics , Retinal Dehydrogenase/metabolism , Signal Transduction , Species Specificity , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/pathology , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/pathology , Vitamin A/administration & dosage
11.
Immunology ; 142(1): 39-45, 2014 May.
Article in English | MEDLINE | ID: mdl-24843870

ABSTRACT

Vitamin A and its active metabolite retinoic acid are essential for the development and function of many tissues including the immune system. The induction of mucosal homing receptors on T and B cells by mucosal dendritic cells (DC) depends on the presence of vitamin A. Recent studies indicate that also the differentiation of CD11b+ DC subsets in the mucosa as well as the spleen depend on vitamin A signalling. As CD11b+ DC subsets exert non-redundant functions in anti-bacterial and anti-fungal immune responses, defects in CD11b+ DC differentiation will contribute to the clinical problems observed during vitamin A deficiency.


Subject(s)
Cell Differentiation/drug effects , Dendritic Cells/drug effects , Vitamin A/pharmacology , Animals , Biomarkers/metabolism , CD11b Antigen/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Immunity, Mucosal/drug effects , Lymphoid Tissue/drug effects , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Vitamin A Deficiency/immunology , Vitamin A Deficiency/metabolism
12.
Immunology ; 142(2): 269-78, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24883436

ABSTRACT

CD169-positive macrophages in the marginal zone of the spleen and subcapsular sinus of lymph nodes play an important role as gatekeepers, strategically located to capture pathogens. Here we identified a population of CD169-positive macrophages in the colon and investigated which factors influenced their development. Murine colonic CD115+ F4/80(lo) CD11c(lo) macrophages expressing CD169 were present in the lamina propria, mainly surrounding the crypts. In spite of the high levels of bacterial flora in the colon and the importance of Toll-like receptor signalling in mucosal homeostasis, the presence of CD169+ macrophages was not affected in mice that were deficient in MyD88-mediated Toll-like receptor signalling and in mice in which the bacterial flora was eradicated. Whereas the development of splenic CD169+ macrophages was dependent on lymphotoxin α, colonic CD169+ macrophages were present in normal numbers in lymphotoxin α-deficient mice. In contrast, reduced numbers of CD169+ macrophages were found in the colon of mice deficient in vitamin A, whereas CD169+ macrophages in the spleen were unaffected. In conclusion, we identified a new macrophage subset in the lamina propria of the colon characterized by the expression of CD169. Its differentiation, unlike CD169+ macrophages in lymphoid organs, is independent of lymphotoxin α signalling, but requires vitamin A.


Subject(s)
Colon/cytology , Colon/immunology , Macrophages/cytology , Macrophages/immunology , Myelopoiesis , Sialic Acid Binding Ig-like Lectin 1/immunology , Animals , Colon/microbiology , Female , Lymphotoxin-alpha , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucous Membrane/cytology , Mucous Membrane/immunology , Vitamin A/metabolism
13.
Eur J Immunol ; 43(6): 1608-16, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23519987

ABSTRACT

The vitamin A metabolite retinoic acid is important for the function of the adaptive immune system, but the mechanism is not completely understood. Here we show that vitamin A is essential for the generation of Notch-dependent CD8(-) dendritic cells (DCs) in the spleen. We observed that CD8(-) CD4(-) (double negative (DN)) and CD4(+) DCs, but not CD8(+) DCs, express vitamin A regulated genes. To determine whether vitamin A levels influence splenic DC development, we generated mice that were fed a vitamin A-deficient diet. We detected a specific reduction of CD4(+) and DN DCs in the spleens of mice fed a vitamin A-deficient diet, while pre-DC numbers in both spleen and bone marrow were not affected. Vitamin A was specifically necessary for the development of RelB(high) , Notch-dependent CD4(+) , and DN DCs. In addition, DN DCs showed reduced proliferation during vitamin A deficiency. In contrast, mice that had received a diet with increased amounts of retinoic acid showed a significant expansion of Notch-dependent DN DCs. These data demonstrate that vitamin A stimulates the development of Notch-dependent splenic DCs and indicate that inefficient generation of DCs may contribute to the immune deficits observed during vitamin A deficiency.


Subject(s)
Dendritic Cells/immunology , Tretinoin/immunology , Vitamin A Deficiency/immunology , Animals , CD4 Antigens/immunology , CD8 Antigens/immunology , Cell Differentiation , Cell Lineage , Cell Proliferation , Feeding Behavior , Female , Mice , Mice, Inbred C57BL , Pregnancy , Receptors, Notch/metabolism , Spleen/cytology , Transcription Factor RelB/metabolism
14.
Proc Natl Acad Sci U S A ; 107(1): 216-21, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-20018690

ABSTRACT

The spleen is the lymphoid organ that induces immune responses toward blood-borne pathogens. Specialized macrophages in the splenic marginal zone are strategically positioned to phagocytose pathogens and cell debris, but are not known to play a role in the activation of T-cell responses. Here we demonstrate that splenic marginal metallophilic macrophages (MMM) are essential for cross-presentation of blood-borne antigens by splenic dendritic cells (DCs). Our data demonstrate that antigens targeted to MMM as well as blood-borne adenoviruses are efficiently captured by MMM and exclusively transferred to splenic CD8(+) DCs for cross-presentation and for the activation of cytotoxic T lymphocytes. Depletion of macrophages in the marginal zone prevents cytotoxic T-lymphocyte activation by CD8(+) DCs after antibody targeting or adenovirus infection. Moreover, we show that tumor antigen targeting to MMM is very effective as antitumor immunotherapy. Our studies point to an important role for splenic MMM in the initial steps of CD8(+) T-cell immunity by capturing and concentrating blood-borne antigens and the transfer to cross-presenting DCs which can be used to design vaccination strategies to induce antitumor cytotoxic T-cell immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Macrophages/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigen Presentation/immunology , Bone Density Conservation Agents/pharmacology , Clodronic Acid/pharmacology , Lymphocyte Activation/immunology , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Spleen/cytology , Spleen/immunology
15.
Front Immunol ; 14: 1290272, 2023.
Article in English | MEDLINE | ID: mdl-38054006

ABSTRACT

Historically platelets are mostly known for their crucial contribution to hemostasis, but there is growing understanding of their role in inflammation and immunity. The immunomodulatory role of platelets entails interaction with pathogens, but also with immune cells including macrophages and dendritic cells (DCs), to activate adaptive immune responses. In our previous work, we have demonstrated that splenic CD169+ macrophages scavenge liposomes and collaborate with conventional type 1 DCs (cDC1) to induce expansion of CD8+ T cells. Here, we show that platelets associate with liposomes and bind to DNGR-1/Clec9a and CD169/Siglec-1 receptors in vitro. In addition, platelets interacted with splenic CD169+ macrophages and cDC1 and further increased liposome internalization by cDC1. Most importantly, platelet depletion prior to liposomal immunization resulted in significantly diminished antigen-specific CD8+ T cell responses, but not germinal center B cell responses. Previously, complement C3 was shown to be essential for platelet-mediated CD8+ T cell activation during bacterial infection. However, after liposomal vaccination CD8+ T cell priming was not dependent on complement C3. While DCs from platelet-deficient mice exhibited unaltered maturation status, they did express lower levels of CCR7. In addition, in the absence of platelets, CCL5 plasma levels were significantly reduced. Overall, our findings demonstrate that platelets engage in a cross-talk with CD169+ macrophages and cDC1 and emphasize the importance of platelets in induction of CD8+ T cell responses in the context of liposomal vaccination.


Subject(s)
CD8-Positive T-Lymphocytes , Liposomes , Animals , Mice , Liposomes/metabolism , Complement C3/metabolism , Macrophages , Antigens
16.
J Immunother Cancer ; 11(11)2023 11.
Article in English | MEDLINE | ID: mdl-37940346

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers. Despite the successful application of immune checkpoint blockade in a range of human cancers, immunotherapy in PDAC remains unsuccessful. PDAC is characterized by a desmoplastic, hypoxic and highly immunosuppressive tumor microenvironment (TME), where T-cell infiltration is often lacking (immune desert), or where T cells are located distant from the tumor islands (immune excluded). Converting the TME to an immune-inflamed state, allowing T-cell infiltration, could increase the success of immunotherapy in PDAC. METHOD: In this study, we use the KPC3 subcutaneous PDAC mouse model to investigate the role of tumor-derived sialic acids in shaping the tumor immune landscape. A sialic acid deficient KPC3 line was generated by genetic knock-out of the CMAS (cytidine monophosphate N-acetylneuraminic acid synthetase) enzyme, a critical enzyme in the synthesis of sialic acid-containing glycans. The effect of sialic acid-deficiency on immunotherapy efficacy was assessed by treatment with anti-programmed cell death protein 1 (PD-1) and agonistic CD40. RESULT: The absence of sialic acids in KPC3 tumors resulted in increased numbers of CD4+ and CD8+ T cells in the TME, and reduced frequencies of CD4+ regulatory T cells (Tregs) within the T-cell population. Importantly, CD8+ T cells were able to infiltrate the tumor islands in sialic acid-deficient tumors. These favorable alterations in the immune landscape sensitized sialic acid-deficient tumors to immunotherapy, which was ineffective in sialic acid-expressing KPC3 tumors. In addition, high expression of sialylation-related genes in human pancreatic cancer correlated with decreased CD8+ T-cell infiltration, increased presence of Tregs, and poorer survival probability. CONCLUSION: Our results demonstrate that tumor-derived sialic acids mediate T-cell exclusion within the PDAC TME, thereby impairing immunotherapy efficacy. Targeting sialic acids represents a potential strategy to enhance T-cell infiltration and improve immunotherapy outcomes in PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Mice , Animals , Humans , CD8-Positive T-Lymphocytes , Sialic Acids/pharmacology , N-Acetylneuraminic Acid/pharmacology , Pancreatic Neoplasms/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Immunotherapy/methods , Tumor Microenvironment
17.
J Immunother Cancer ; 10(7)2022 07.
Article in English | MEDLINE | ID: mdl-35853671

ABSTRACT

BACKGROUND: Many solid tumors do not respond to immunotherapy due to their immunologically cold tumor microenvironment (TME). We and others found that oncolytic viruses (OVs), including reovirus type 3 Dearing, can enhance the efficacy of immunotherapy by recruiting CD8+ T cells to the TME. A significant part of the incoming CD8+ T cells is directed toward reovirus itself, which may be detrimental to the efficacy of OVs. However, here we aim to exploit these incoming virus-specific T cells as anticancer effector cells. METHODS: We performed an in-depth characterization of the reovirus-induced T-cell response in immune-competent mice bearing pancreatic KPC3 tumors. The immunodominant CD8+ T-cell epitope of reovirus was identified using epitope prediction algorithms and peptide arrays, and the quantity and quality of reovirus-specific T cells after reovirus administration were assessed using high-dimensional flow cytometry. A synthetic long peptide (SLP)-based vaccination strategy was designed to enhance the intratumoral frequency of reovirus-specific CD8+ T cells. RESULTS: Reovirus administration did not induce tumor-specific T cells but rather induced high frequencies of reovirus-specific CD8+ T cells directed to the immunodominant epitope. Priming of reovirus-specific T cells required a low-frequent population of cross-presenting dendritic cells which was absent in Batf3-/- mice. While intratumoral and intravenous reovirus administration induced equal systemic frequencies of reovirus-specific T cells, reovirus-specific T cells were highly enriched in the TME exclusively after intratumoral administration. Here, they displayed characteristics of potent effector cells with high expression of KLRG1, suggesting they may be responsive against local reovirus-infected cells. To exploit these reovirus-specific T cells as anticancer effector cells, we designed an SLP-based vaccination strategy to induce a strong T-cell response before virotherapy. These high frequencies of circulating reovirus-specific T cells were reactivated on intratumoral reovirus administration and significantly delayed tumor growth. CONCLUSIONS: These findings provide proof of concept that OV-specific T cells, despite not being tumor-specific, can be exploited as potent effector cells for anticancer treatment when primed before virotherapy. This is an attractive strategy for low-immunogenic tumors lacking tumor-specific T cells.


Subject(s)
Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Animals , CD8-Positive T-Lymphocytes , Immunotherapy , Mice , Oncolytic Virotherapy/methods , Tumor Microenvironment
18.
Front Immunol ; 13: 842241, 2022.
Article in English | MEDLINE | ID: mdl-35251040

ABSTRACT

Cancer vaccination aims to activate immunity towards cancer cells and can be achieved by delivery of cancer antigens together with immune stimulatory adjuvants to antigen presenting cells (APC). APC maturation and antigen processing is a subsequent prerequisite for T cell priming and anti-tumor immunity. In order to specifically target APC, nanoparticles, such as liposomes, can be used for the delivery of antigen and adjuvant. We have previously shown that liposomal inclusion of the ganglioside GM3, an endogenous ligand for CD169, led to robust uptake by CD169-expressing APC and resulted in strong immune responses when supplemented with a soluble adjuvant. To minimize the adverse effects related to a soluble adjuvant, immune stimulatory molecules can be incorporated in liposomes to achieve targeted delivery of both antigen and adjuvant. In this study, we incorporated TLR4 (MPLA) or TLR7/8 (3M-052) ligands in combination with inflammasome stimuli, 1-palmitoyl-2-glutaryl-sn-glycero-3-phosphocholine (PGPC) or muramyl dipeptide (MDP), into GM3 liposomes. Incorporation of TLR and inflammasome ligands did not interfere with the uptake of GM3 liposomes by CD169-expressing cells. GM3 liposomes containing a TLR ligand efficiently matured human and mouse dendritic cells in vitro and in vivo, while inclusion of PGPC or MDP had minor effects on maturation. Immunization with MPLA-containing GM3 liposomes containing an immunogenic synthetic long peptide stimulated CD4+ and CD8+ T cell responses, but additional incorporation of either PGPC or MDP did not translate into stronger immune responses. In conclusion, our study indicates that TLRL-containing GM3 liposomes are effective vectors to induce DC maturation and T cell priming and thus provide guidance for further selection of liposomal components to optimally stimulate anti-cancer immune responses.


Subject(s)
Liposomes , Neoplasms , Adjuvants, Immunologic/pharmacology , Animals , Antigens/metabolism , Dendritic Cells , Inflammasomes/metabolism , Ligands , Liposomes/chemistry , Mice , Toll-Like Receptors/metabolism
19.
J Exp Med ; 219(2)2022 02 07.
Article in English | MEDLINE | ID: mdl-34919143

ABSTRACT

Tumor-associated macrophages (TAMs) are correlated with the progression of prostatic adenocarcinoma (PCa). The mechanistic basis of this correlation and therapeutic strategies to target TAMs in PCa remain poorly defined. Here, single-cell RNA sequencing was used to profile the transcriptional landscape of TAMs in human PCa, leading to identification of a subset of macrophages characterized by dysregulation in transcriptional pathways associated with lipid metabolism. This subset of TAMs correlates positively with PCa progression and shorter disease-free survival and is characterized by an accumulation of lipids that is dependent on Marco. Mechanistically, cancer cell-derived IL-1ß enhances Marco expression on macrophages, and reciprocally, cancer cell migration is promoted by CCL6 released by lipid-loaded TAMs. Moreover, administration of a high-fat diet to tumor-bearing mice raises the abundance of lipid-loaded TAMs. Finally, targeting lipid accumulation by Marco blockade hinders tumor growth and invasiveness and improves the efficacy of chemotherapy in models of PCa, pointing to combinatorial strategies that may influence patient outcomes.


Subject(s)
Lipids , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , Tumor Microenvironment , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Animals , Cell Plasticity/genetics , Cell Plasticity/immunology , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Gene Expression Profiling , Gene Knockdown Techniques , Heterografts , Humans , Lipid Metabolism , Lipids/chemistry , Male , Metabolic Networks and Pathways , Mice , Prostatic Neoplasms/pathology , Single-Cell Analysis
20.
Pharmaceutics ; 13(7)2021 Jun 24.
Article in English | MEDLINE | ID: mdl-34202919

ABSTRACT

Liposomes have emerged as interesting vehicles in cancer vaccination strategies as their composition enables the inclusion of both hydrophilic and hydrophobic antigens and adjuvants. In addition, liposomes can be decorated with targeting moieties to further resemble pathogenic particles that allow for better engagement with the immune system. However, so far liposomal cancer vaccines have not yet reached their full potential in the clinic. In this review, we summarize recent preclinical studies on liposomal cancer vaccines. We describe the basic ingredients for liposomal cancer vaccines, tumor antigens, and adjuvants, and how their combined inclusion together with targeting moieties potentially derived from pathogens can enhance vaccine immunogenicity. We discuss newly identified antigen-presenting cells in humans and mice that pose as promising targets for cancer vaccines. The lessons learned from these preclinical studies can be applied to enhance the efficacy of liposomal cancer vaccination in the clinic.

SELECTION OF CITATIONS
SEARCH DETAIL