Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 183(2): 411-428.e16, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32970988

ABSTRACT

The colon is primarily responsible for absorbing fluids. It contains a large number of microorganisms including fungi, which are enriched in its distal segment. The colonic mucosa must therefore tightly regulate fluid influx to control absorption of fungal metabolites, which can be toxic to epithelial cells and lead to barrier dysfunction. How this is achieved remains unknown. Here, we describe a mechanism by which the innate immune system allows rapid quality check of absorbed fluids to avoid intoxication of colonocytes. This mechanism relies on a population of distal colon macrophages that are equipped with "balloon-like" protrusions (BLPs) inserted in the epithelium, which sample absorbed fluids. In the absence of macrophages or BLPs, epithelial cells keep absorbing fluids containing fungal products, leading to their death and subsequent loss of epithelial barrier integrity. These results reveal an unexpected and essential role of macrophages in the maintenance of colon-microbiota interactions in homeostasis. VIDEO ABSTRACT.


Subject(s)
Gastrointestinal Microbiome/physiology , Intestinal Mucosa/metabolism , Macrophages/metabolism , Animals , Colon/metabolism , Epithelial Cells/metabolism , Epithelium , Female , Homeostasis , Immunity, Innate/immunology , Intestinal Mucosa/microbiology , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Microbiota , Signal Transduction
2.
Immunity ; 50(6): 1453-1466.e4, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31053503

ABSTRACT

In lymph nodes, subcapsular sinus macrophages (SSMs) form an immunological barrier that monitors lymph drained from peripheral tissues. Upon infection, SSMs activate B and natural killer T (NKT) cells while secreting inflammatory mediators. Here, we investigated the mechanisms regulating development and homeostasis of SSMs. Embryonic SSMs originated from yolk sac hematopoiesis and were replaced by a postnatal wave of bone marrow (BM)-derived monocytes that proliferated to establish the adult SSM network. The SSM network self-maintained by proliferation with minimal BM contribution. Upon pathogen-induced transient deletion, BM-derived cells contributed to restoring the SSM network. Lymphatic endothelial cells (LECs) were the main source of CSF-1 within the lymph node and conditional deletion of Csf1 in adult LECs decreased the network of SSMs and medullary sinus macrophages (MSMs). Thus, SSMs have a dual hematopoietic origin, and LECs are essential to the niche supporting these macrophages.


Subject(s)
Endothelial Cells/metabolism , Macrophages/metabolism , Animals , Biomarkers , Cell Communication , Cell Differentiation , Gene Expression , Genes, Reporter , Hematopoiesis/genetics , Hematopoiesis/immunology , Homeostasis , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphatic Vessels , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/cytology , Macrophages/immunology , Mice , Monocytes/cytology , Monocytes/metabolism , Yolk Sac
3.
Immunity ; 49(2): 312-325.e5, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30076102

ABSTRACT

Heterogeneity between different macrophage populations has become a defining feature of this lineage. However, the conserved factors defining macrophages remain largely unknown. The transcription factor ZEB2 is best described for its role in epithelial to mesenchymal transition; however, its role within the immune system is only now being elucidated. We show here that Zeb2 expression is a conserved feature of macrophages. Using Clec4f-cre, Itgax-cre, and Fcgr1-cre mice to target five different macrophage populations, we found that loss of ZEB2 resulted in macrophage disappearance from the tissues, coupled with their subsequent replenishment from bone-marrow precursors in open niches. Mechanistically, we found that ZEB2 functioned to maintain the tissue-specific identities of macrophages. In Kupffer cells, ZEB2 achieved this by regulating expression of the transcription factor LXRα, removal of which recapitulated the loss of Kupffer cell identity and disappearance. Thus, ZEB2 expression is required in macrophages to preserve their tissue-specific identities.


Subject(s)
Kupffer Cells/cytology , Liver X Receptors/genetics , Zinc Finger E-box Binding Homeobox 2/genetics , Animals , Cell Lineage/immunology , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Kupffer Cells/immunology , Liver/cytology , Liver X Receptors/metabolism , Lung/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
4.
Nat Immunol ; 15(10): 929-937, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25151491

ABSTRACT

The paradigm that macrophages that reside in steady-state tissues are derived from embryonic precursors has never been investigated in the intestine, which contains the largest pool of macrophages. Using fate-mapping models and monocytopenic mice, together with bone marrow chimera and parabiotic models, we found that embryonic precursor cells seeded the intestinal mucosa and demonstrated extensive in situ proliferation during the neonatal period. However, these cells did not persist in the intestine of adult mice. Instead, they were replaced around the time of weaning by the chemokine receptor CCR2-dependent influx of Ly6C(hi) monocytes that differentiated locally into mature, anti-inflammatory macrophages. This process was driven largely by the microbiota and had to be continued throughout adult life to maintain a normal intestinal macrophage pool.


Subject(s)
Intestinal Mucosa/immunology , Intestines/immunology , Macrophages/immunology , Monocytes/immunology , Animals , Animals, Newborn , Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , Antigens, Differentiation/metabolism , Antigens, Ly/immunology , Antigens, Ly/metabolism , Bone Marrow Transplantation , CD11b Antigen/genetics , CD11b Antigen/immunology , CD11b Antigen/metabolism , CX3C Chemokine Receptor 1 , Cell Differentiation/immunology , Cell Proliferation , Flow Cytometry , Gene Expression/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestines/cytology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Immunological , Monocytes/metabolism , Parabiosis , Receptors, CCR2/genetics , Receptors, CCR2/immunology , Receptors, CCR2/metabolism , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
5.
Immunity ; 45(2): 305-18, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27533013

ABSTRACT

Dendritic cells (DCs) are instrumental in the initiation of T cell responses, but how thymic and peripheral tolerogenic DCs differ globally from Toll-like receptor (TLR)-induced immunogenic DCs remains unclear. Here, we show that thymic XCR1(+) DCs undergo a high rate of maturation, accompanied by profound gene-expression changes that are essential for central tolerance and also happen in germ-free mice. Those changes largely overlap those occurring during tolerogenic and, more unexpectedly, TLR-induced maturation of peripheral XCR1(+) DCs, arguing against the commonly held view that tolerogenic DCs undergo incomplete maturation. Interferon-stimulated gene (ISG) expression was among the few discriminators of immunogenic and tolerogenic XCR1(+) DCs. Tolerogenic XCR1(+) thymic DCs were, however, unique in expressing ISGs known to restrain virus replication. Therefore, a broad functional convergence characterizes tolerogenic and immunogenic XCR1(+) DC maturation in the thymus and periphery, maximizing antigen presentation and signal delivery to developing and to conventional and regulatory mature T cells.


Subject(s)
Central Tolerance , Dendritic Cells/immunology , Peripheral Tolerance , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Animals , Antigen Presentation , Cell Differentiation , Cells, Cultured , Interferon Regulatory Factors/genetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Chemokine/metabolism , Toll-Like Receptors/immunology , Transcriptome , Virus Replication
6.
Immunity ; 45(3): 669-684, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27637149

ABSTRACT

Dendritic cells (DCs) are professional antigen-presenting cells that hold great therapeutic potential. Multiple DC subsets have been described, and it remains challenging to align them across tissues and species to analyze their function in the absence of macrophage contamination. Here, we provide and validate a universal toolbox for the automated identification of DCs through unsupervised analysis of conventional flow cytometry and mass cytometry data obtained from multiple mouse, macaque, and human tissues. The use of a minimal set of lineage-imprinted markers was sufficient to subdivide DCs into conventional type 1 (cDC1s), conventional type 2 (cDC2s), and plasmacytoid DCs (pDCs) across tissues and species. This way, a large number of additional markers can still be used to further characterize the heterogeneity of DCs across tissues and during inflammation. This framework represents the way forward to a universal, high-throughput, and standardized analysis of DC populations from mutant mice and human patients.


Subject(s)
Dendritic Cells/physiology , Animals , Cell Differentiation/physiology , Flow Cytometry , Humans , Inflammation/pathology , Macaca , Mice , Mice, Inbred C57BL
7.
PLoS Pathog ; 17(7): e1009749, 2021 07.
Article in English | MEDLINE | ID: mdl-34324611

ABSTRACT

The main avenue for the development of an HIV-1 vaccine remains the induction of protective antibodies. A rationale approach is to target antigen to specific receptors on dendritic cells (DC) via fused monoclonal antibodies (mAb). In mouse and non-human primate models, targeting of skin Langerhans cells (LC) with anti-Langerin mAbs fused with HIV-1 Gag antigen drives antigen-specific humoral responses. The development of these immunization strategies in humans requires a better understanding of early immune events driven by human LC. We therefore produced anti-Langerin mAbs fused with the HIV-1 gp140z Envelope (αLC.Env). First, we show that primary skin human LC and in vitro differentiated LC induce differentiation and expansion of naïve CD4+ T cells into T follicular helper (Tfh) cells. Second, when human LC are pre-treated with αLC.Env, differentiated Tfh cells significantly promote the production of specific IgG by B cells. Strikingly, HIV-Env-specific Ig are secreted by HIV-specific memory B cells. Consistently, we found that receptors and cytokines involved in Tfh differentiation and B cell functions are upregulated by LC during their maturation and after targeting Langerin. Finally, we show that subcutaneous immunization of mice by αLC.Env induces germinal center (GC) reaction in draining lymph nodes with higher numbers of Tfh cells, Env-specific B cells, as well as specific IgG serum levels compared to mice immunized with the non-targeting Env antigen. Altogether, we provide evidence that human LC properly targeted may be licensed to efficiently induce Tfh cell and B cell responses in GC.


Subject(s)
AIDS Vaccines/immunology , Antigens, CD/immunology , HIV-1/immunology , Immunity, Humoral/immunology , Langerhans Cells/immunology , Lectins, C-Type/immunology , Mannose-Binding Lectins/immunology , Animals , Humans , Lymphocyte Activation/immunology , Mice , env Gene Products, Human Immunodeficiency Virus/immunology
8.
EMBO Rep ; 22(4): e52196, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33719206

ABSTRACT

T and B cells continually recirculate between blood and secondary lymphoid organs. To promote their trans-endothelial migration (TEM), chemokine receptors control the activity of RHO family small GTPases in part via GTPase-activating proteins (GAPs). T and B cells express several RHO-GAPs, the function of most of which remains unknown. The ARHGAP45 GAP is predominantly expressed in hematopoietic cells. To define its in vivo function, we describe two mouse models where ARHGAP45 is ablated systemically or selectively in T cells. We combine their analysis with affinity purification coupled to mass spectrometry to determine the ARHGAP45 interactome in T cells and with time-lapse and reflection interference contrast microscopy to assess the role of ARGHAP45 in T-cell polarization and motility. We demonstrate that ARHGAP45 regulates naïve T-cell deformability and motility. Under physiological conditions, ARHGAP45 controls the entry of naïve T and B cells into lymph nodes whereas under competitive repopulation it further regulates hematopoietic progenitor cell engraftment in the bone marrow, and T-cell progenitor thymus seeding. Therefore, the ARGHAP45 GAP controls multiple key steps in the life of T and B cells.


Subject(s)
T-Lymphocytes , Virus Internalization , Animals , B-Lymphocytes , Cell Movement , GTPase-Activating Proteins/genetics , Lymph Nodes , Mice , Thymus Gland
9.
J Allergy Clin Immunol ; 150(5): 1194-1208, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35779666

ABSTRACT

BACKGROUND: Epicutaneous immunotherapy (EPIT) protocols have recently been developed to restore tolerance in patients with food allergy. The mechanisms by which EPIT protocols promote desensitization rely on a profound immune deviation of pathogenic T- and B-cell responses. OBJECTIVE: To date, little is known about the contribution of skin dendritic cells (skDCs) to T-cell remodeling and EPIT efficacy. METHODS: We capitalized on a preclinical model of food allergy to ovalbumin (OVA) to characterize the phenotype and functions of OVA+ skDCs throughout the course of EPIT. RESULTS: Our results showed that both Langerhans cells and dermal conventional cDC1 and cDC2 subsets retained their ability to capture OVA in the skin and to migrate toward the skin-draining lymph nodes during EPIT. However, their activation/maturation status was significantly impaired, as evidenced by the gradual and selective reduction of CD86, CD40, and OVA protein expression in respective subsets. Phenotypic changes during EPIT were also characterized by a progressive diversification of single-cell gene signatures within each DC subset. Interestingly, we observed that OVA+ Langerhans cells progressively lost their capacity to prime CD4+ TEFF cells, but gained regulatory T-cell stimulatory properties. In contrast, cDC1 were inefficient in priming CD4+ TEFF cells or in reactivating TMEM cells in vitro, whereas cDC2 retained moderate stimulatory properties, and progressively biased type 2 immunity toward type 1 and type 17 responses. CONCLUSIONS: Our results therefore emphasize that the acquisition of distinct phenotypic and functional specializations by skDCs during EPIT is at the cornerstone of the desensitization process.


Subject(s)
Food Hypersensitivity , Langerhans Cells , Humans , Desensitization, Immunologic/methods , Ovalbumin , T-Lymphocytes, Regulatory , Allergens
10.
Immunity ; 39(5): 925-38, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24184057

ABSTRACT

In the skin, the lack of markers permitting the unambiguous identification of macrophages and of conventional and monocyte-derived dendritic cells (DCs) complicates understanding of their contribution to skin integrity and to immune responses. By combining CD64 and CCR2 staining, we successfully identified each of these cell types and studied their origin, transcriptomic signatures, and migratory and T cell stimulatory properties. We also analyzed the impact of microbiota on their development and their contribution to skin inflammation during contact hypersensitivity. Dermal macrophages had a unique scavenging role and were unable to migrate and activate T cells. Conventional dermal DCs excelled both at migrating and activating T cells. In the steady-state dermis, monocyte-derived DCs are continuously generated by extravasated Ly-6C(hi) monocytes. Their T cell stimulatory capacity combined with their poor migratory ability made them particularly suited to activate skin-tropic T cells. Therefore, a high degree of functional specialization occurs among the mononuclear phagocytes of the skin.


Subject(s)
Dendritic Cells/cytology , Macrophages/cytology , Skin/cytology , Animals , Antigens, Differentiation/analysis , CD11b Antigen/analysis , Cell Lineage , Chemotaxis, Leukocyte , Chromatography, Gel , Dendritic Cells/immunology , Dermatitis, Contact/immunology , Dermatitis, Contact/pathology , Dermis/cytology , Gene Expression Regulation, Developmental , Immunophenotyping/methods , Langerhans Cells/cytology , Langerhans Cells/immunology , Lymphocyte Cooperation , Macrophages/physiology , Mice , Microbiota/immunology , Monocytes/cytology , Principal Component Analysis , Radiation Chimera , Receptors, CCR2/analysis , Receptors, IgG/analysis , Skin/immunology , Skin/microbiology , Specific Pathogen-Free Organisms , Staining and Labeling/methods , Transcriptome
11.
Dermatology ; 237(2): 296-302, 2021.
Article in English | MEDLINE | ID: mdl-32344413

ABSTRACT

BACKGROUND: Little information is available about the complexity and function of skin cells contributing to the high stability of tattoos. It has been shown that dermal macrophages play an important role in the storage and maintenance of pigment particles. By contrast, the impact of dermal fibroblasts, forming the connective tissue of the skin, on the stability of the tattoo is not known. METHOD: In this study, we compared the cell number and the particle load in dermal macrophages versus dermal fibroblasts, isolated from tail skin of tattooed mice. RESULTS: Microscopic analysis revealed that both cell populations contained the tattoo particles, although in largely different amounts. A small number of macrophages with high side scatter intensity contained a large quantity of pigment particles, whereas a high number of dermal fibroblasts harbored only a few pigment particles. Using the CD64dtr mouse model that allows for selective, diphtheria toxin-mediated depletion of macrophages, we have previously shown that macrophages hold the tattoo in place by capture-release and recapture cycles. In the tattooed skin of macrophage-depleted mice, the content of pigment particles in fibroblasts did not change; however, the total number of fibroblasts carrying particles increased. CONCLUSION: The present study demonstrates that dermal macrophages and fibroblasts contribute in different ways to the tattoo stability and further improves our knowledge on tattoo persistence.


Subject(s)
Coloring Agents , Dermis/cytology , Fibroblasts/physiology , Macrophages/physiology , Tattooing , Animals , Cell Count , Ink , Mice , Microscopy
12.
Nanomedicine ; 32: 102321, 2021 02.
Article in English | MEDLINE | ID: mdl-33184020

ABSTRACT

Ultra-small 1-2 nm gold nanoparticles (NP) were conjugated with a poorly-soluble peptide auto-antigen, associated with type 1 diabetes, to modify the peptide pharmacokinetics, following its intradermal delivery. Peptide distribution was characterized, in vivo, after delivery using either conventional intradermal injection or a hollow microneedle device. The poorly-soluble peptide was effectively presented in distant lymph nodes (LN), spleen and draining LN when conjugated to the nanoparticles, whereas peptide alone was only presented in the draining LN. By contrast, nanoparticle conjugation to a highly-soluble peptide did not enhance in vivo distribution. Transfer of both free peptide and peptide-NPs from the skin to LN was reduced in mice lacking lymphoid homing receptor CCR7, suggesting that both are actively transported by migrating dendritic cells to LN. Collectively, these data demonstrate that intradermally administered ultra-small gold nanoparticles can widen the distribution of poorly-soluble auto-antigenic peptides to multiple lymphoid organs, thus enhancing their use as potential therapeutics.


Subject(s)
Antigens/metabolism , Gold/chemistry , Metal Nanoparticles/chemistry , Peptides/pharmacology , Amino Acid Sequence , Animals , Cell Proliferation , Dendritic Cells/drug effects , Injections, Intradermal , Mice, Inbred C57BL , Mice, Transgenic , Needles , Peptides/chemistry , Peptides/pharmacokinetics , Phenotype , Skin/drug effects , Solubility , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
13.
Article in English | MEDLINE | ID: mdl-32638670

ABSTRACT

OBJECTIVES: Unlike randomized controlled trials, lack of methodological rigor is a concern about real-world evidence (RWE) studies. The objective of this study was to characterize methodological practices of studies collecting pharmacoeconomic data in a real-world setting for the management of type 2 diabetes mellitus (T2DM). METHODS: A systematic literature review was performed using the PICO framework: population consisted of T2DM patients, interventions and comparators were any intervention for T2DM care or absence of intervention, and outcomes were resource utilization, productivity loss or utility. Only RWE studies were included, defined as studies that were not clinical trials and that collected de novo data (no retrospective analysis). RESULTS: The literature search identified 1,158 potentially relevant studies, among which sixty were included in the literature review. Many studies showed a lack of transparency by not mentioning the source for outcome and exposure measurement, source for patient selection, number of study sites, recruitment duration, sample size calculation, sampling method, missing data, approbation by an ethics committee, obtaining patient's consent, conflicts of interest, and funding. A significant proportion of studies had poor quality scores and was at high risk of bias. CONCLUSIONS: RWE from T2DM studies lacks transparency and credibility. There is a need for good procedural practices that can increase confidence in RWE studies. Standardized methodologies specifically adapted for RWE studies collecting pharmacoeconomic data for the management of T2DM could help future reimbursement decision making in this major public health problem.

14.
Immunol Rev ; 262(1): 9-24, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25319324

ABSTRACT

Tissues that are in direct contact with the outside world face particular immunological challenges. The intestine, the skin, and the lung possess important mononuclear phagocyte populations to deal with these challenges, but the cellular origin of these phagocytes is strikingly different from one subset to another, with some cells derived from embryonic precursors and some from bone marrow-derived circulating monocytes. Here, we review the current knowledge regarding the developmental pathways that control the differentiation of mononuclear phagocytes in these barrier tissues. We have also attempted to build a theoretical model that could explain the distinct cellular origin of mononuclear phagocytes in these tissues.


Subject(s)
Intestines/physiology , Lung/physiology , Mononuclear Phagocyte System/cytology , Mononuclear Phagocyte System/physiology , Skin Physiological Phenomena , Animals , Cell Differentiation , Cell Movement , Cellular Microenvironment , Humans , Models, Biological , Organ Specificity
15.
J Immunol ; 195(10): 4953-61, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26466959

ABSTRACT

Psoriasis is a chronic inflammatory skin disease of unknown etiology. Previous studies showed that short-term, 5-7 d-long application of imiquimod (IMQ), a TLR7 agonist, to the skin of mice triggers a psoriasis-like inflammation. In the current study, by applying IMQ for 14 consecutive d, we established an improved mouse psoriasis-like model in that it recapitulated many of the clinical and cellular hallmarks observed in human patients during both the early-onset and the late-stable phase of psoriasis. Although macrophages and dendritic cells (DCs) have been proposed to drive the psoriatic cascade, their largely overlapping phenotype hampered studying their respective role. Based on our ability to discriminate Langerhans cells (LCs), conventional DCs, monocytes, monocyte-derived DCs, macrophages, and plasmacytoid DCs in the skin, we addressed their dynamics during both phases of our biphasic psoriasis-like model. Plasmacytoid DCs were not detectable during the whole course of IMQ treatment. During the early phase, neutrophils infiltrated the epidermis, whereas monocytes and monocyte-derived DCs were predominant in the dermis. During the late phase, LCs and macrophage numbers transiently increased in the epidermis and dermis, respectively. LC expansion resulted from local proliferation, a conclusion supported by global transcriptional analysis. Genetic depletion of LCs permitted to evaluate their function during both phases of the biphasic psoriasis-like model and demonstrated that their absence resulted in a late phase that is associated with enhanced neutrophil infiltration. Therefore, our data support an anti-inflammatory role of LCs during the course of psoriasis-like inflammation.


Subject(s)
Aminoquinolines/adverse effects , Dendritic Cells/immunology , Macrophages/immunology , Psoriasis/chemically induced , Psoriasis/immunology , Skin/immunology , Transcriptome/drug effects , Aminoquinolines/pharmacology , Animals , Dendritic Cells/pathology , Disease Models, Animal , Humans , Imiquimod , Macrophages/pathology , Mice , Mice, Transgenic , Psoriasis/pathology , Skin/pathology
16.
J Immunol ; 194(12): 5895-902, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25941327

ABSTRACT

The development of vaccines inducing efficient CD8(+) T cell responses is the focus of intense research. Dendritic cells (DCs) expressing the XCR1 chemokine receptor, also known as CD103(+) or CD8α(+) DCs, excel in the presentation of extracellular Ags to CD8(+) T cells. Because of its high numbers of DCs, including XCR1(+) DCs, the skin dermis is an attractive site for vaccine administration. By creating laser-generated micropores through the epidermis, we targeted a model protein Ag fused to XCL1, the ligand of XCR1, to dermal XCR1(+) DCs and induced Ag-specific CD8(+) and CD4(+) T cell responses. Efficient immunization required the emigration of XCR1(+) dermal DCs to draining lymph nodes and occurred irrespective of TLR signaling. Moreover, a single intradermal immunization protected mice against melanoma tumor growth in prophylactic and therapeutic settings, in the absence of exogenous adjuvant. The mild inflammatory milieu created in the dermis by skin laser microporation itself most likely favored the development of potent T cell responses in the absence of exogenous adjuvants. The existence of functionally equivalent XCR1(+) dermal DCs in humans should permit the translation of laser-assisted intradermal delivery of a tumor-specific vaccine targeting XCR1(+) DCs to human cancer immunotherapy. Moreover, considering that the use of adjuvants in vaccines is often associated with safety issues, the possibility of inducing protective responses against melanoma tumor growth independently of the administration of exogenous adjuvants should facilitate the development of safer vaccines.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Neoplasms/immunology , Receptors, Chemokine/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cancer Vaccines/administration & dosage , Chemokines, C/genetics , Chemokines, C/metabolism , Disease Models, Animal , Injections, Intradermal , Melanoma, Experimental , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , Neoplasms/pathology , Neoplasms/therapy , Ovalbumin/genetics , Ovalbumin/immunology , Protein Binding , Receptors, Chemokine/genetics , T-Lymphocyte Subsets/immunology , Tumor Burden/immunology
17.
Proc Natl Acad Sci U S A ; 111(4): 1497-502, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24474776

ABSTRACT

Systemic lupus erythematosus (SLE) is a complex autoimmune disease with diverse clinical presentations characterized by the presence of autoantibodies to nuclear components. Toll-like receptor (TLR)7, TLR8, and TLR9 sense microbial or endogenous nucleic acids and are implicated in the development of SLE. In mice TLR7-deficiency ameliorates SLE, but TLR8- or TLR9-deficiency exacerbates the disease because of increased TLR7 response. Thus, both TLR8 and TLR9 control TLR7 function, but whether TLR8 and TLR9 act in parallel or in series in the same or different cell types in controlling TLR7-mediated lupus remains unknown. Here, we reveal that double TLR8/9-deficient (TLR8/9(-/-)) mice on the C57BL/6 background showed increased abnormalities characteristic of SLE, including splenomegaly, autoantibody production, frequencies of marginal zone and B1 B cells, and renal pathology compared with single TLR8(-/-) or TLR9(-/-) mice. On the cellular level, TLR8(-/-) and TLR8/9(-/-) dendritic cells were hyperesponsive to TLR7 ligand R848, but TLR9(-/-) cells responded normally. Moreover, B cells from TLR9(-/-) and TLR8/9(-/-) mice were hyperesponsive to R848, but TLR8(-/-) B cells were not. These results reveal that TLR8 and TLR9 have an additive effect on controlling TLR7 function and TLR7-mediated lupus; however, they act on different cell types. TLR8 controls TLR7 function on dendritic cells, and TLR9 restrains TLR7 response on B cells.


Subject(s)
Autoimmunity/physiology , B-Lymphocytes/immunology , Dendritic Cells/immunology , Membrane Glycoproteins/physiology , Toll-Like Receptor 7/physiology , Toll-Like Receptor 8/physiology , Toll-Like Receptor 9/physiology , Animals , Flow Cytometry , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Toll-Like Receptor 7/genetics , Toll-Like Receptor 8/genetics , Toll-Like Receptor 9/genetics
18.
Eur J Immunol ; 45(2): 624-35, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25410055

ABSTRACT

Targeting antigens to cross-presenting dendritic cells (DCs) is a promising method for enhancing CD8(+) T-cell responses. However, expression patterns of surface receptors often vary between species, making it difficult to relate observations in mice to other animals. Recent studies have indicated that the chemokine receptor Xcr1 is selectively expressed on cross-presenting murine CD8α(+) DCs, and that the expression is conserved on homologous DC subsets in humans (CD141(+) DCs), sheep (CD26(+) DCs), and macaques (CADM1(+) DCs). We therefore tested if targeting antigens to Xcr1 on cross-presenting DCs using antigen fused to Xcl1, the only known ligand for Xcr1, could enhance immune responses. Bivalent Xcl1 fused to model antigens specifically bound CD8α(+) DCs and increased proliferation of antigen-specific T cells. DNA vaccines encoding dimeric Xcl1-hemagglutinin (HA) fusion proteins induced cytotoxic CD8(+) T-cell responses, and mediated full protection against a lethal challenge with influenza A virus. In addition to enhanced CD8(+) T-cell responses, targeting of antigen to Xcr1 induced CD4(+) Th1 responses and highly selective production of IgG2a antibodies. In conclusion, targeting of dimeric fusion vaccine molecules to CD8α(+) DCs using Xcl1 represents a novel and promising method for induction of protective CD8(+) T-cell responses.


Subject(s)
Antibodies, Viral/biosynthesis , Dendritic Cells/drug effects , Immunoglobulin G/biosynthesis , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Receptors, Chemokine/immunology , T-Lymphocytes, Cytotoxic/drug effects , Animals , Cell Proliferation , Cross-Priming , Dendritic Cells/immunology , Female , Gene Expression , HEK293 Cells , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Immunity, Cellular , Influenza A virus/drug effects , Influenza A virus/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Transgenic , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Receptors, Chemokine/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/drug effects , Th1 Cells/immunology
19.
J Immunol ; 188(4): 1751-60, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22262658

ABSTRACT

Although most vaccines are administered i.m., little is known about the dendritic cells (DCs) that are present within skeletal muscles. In this article, we show that expression of CD64, the high-affinity IgG receptor FcγRI, distinguishes conventional DCs from monocyte-derived DCs (Mo-DCs). By using such a discriminatory marker, we defined the distinct DC subsets that reside in skeletal muscles and identified their migratory counterparts in draining lymph nodes (LNs). We further used this capability to analyze the functional specialization that exists among muscle DCs. After i.m. administration of Ag adsorbed to alum, we showed that alum-injected muscles contained large numbers of conventional DCs that belong to the CD8α(+)- and CD11b(+)-type DCs. Both conventional DC types were capable of capturing Ag and of migrating to draining LNs, where they efficiently activated naive T cells. In alum-injected muscles, Mo-DCs were as numerous as conventional DCs, but only a small fraction migrated to draining LNs. Therefore, alum by itself poorly induces Mo-DCs to migrate to draining LNs. We showed that addition of small amounts of LPS to alum enhanced Mo-DC migration. Considering that migratory Mo-DCs had, on a per cell basis, a higher capacity to induce IFN-γ-producing T cells than conventional DCs, the addition of LPS to alum enhanced the overall immunogenicity of Ags presented by muscle-derived DCs. Therefore, a full understanding of the role of adjuvants during i.m. vaccination needs to take into account the heterogeneous migratory and functional behavior of muscle DCs and Mo-DCs revealed in this study.


Subject(s)
Dendritic Cells/immunology , Monocytes/immunology , Muscle, Skeletal/immunology , Receptors, IgG/biosynthesis , Adjuvants, Immunologic , Alum Compounds , Animals , Cell Differentiation , Immunization , Injections, Intramuscular , Interferon-gamma/biosynthesis , Mice , Receptors, IgG/metabolism , T-Lymphocytes/immunology , Vaccines/administration & dosage , Vaccines/immunology
20.
Immunol Rev ; 234(1): 177-98, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20193019

ABSTRACT

During evolution, vertebrates have developed an adaptive immune system able to cope with a variety of pathogens. Dendritic cells (DCs) are central to this process. DCs integrate information derived from pathogens or endogenous danger signals and convey them to T lymphocytes. Most of the present knowledge on DCs was generated in mice or by using human DCs differentiated in vitro from monocytes. In both species, several DC subsets have been identified in vivo based on differences in their phenotypes, anatomical locations or functions. In mice, protective immunity against intracellular pathogens or tumors can be induced most efficiently by targeting antigens to the CD8 alpha(+) DCs, a subset of DCs which resides in lymphoid tissues and is especially efficient at cross-presenting exogenous antigens to CD8(+) T lymphocytes. In contrary, harnessing human DC subsets for medical purposes is currently hampered by insufficient knowledge about these cells. To overcome this cognitive gap, we are using comparative genomics as a tool for designing hypotheses and experiments to further characterize DC subset functions and their molecular control, including the investigation of the functional equivalences that might exist between human and mouse DC subsets.


Subject(s)
Cell Differentiation , Cell Lineage , Dendritic Cells/immunology , Genomics , Animals , Antigens, CD1 , Antigens, Surface/immunology , CD11b Antigen/immunology , CD8 Antigens/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Communicable Diseases/immunology , Gene Expression Profiling , Genomics/methods , Glycoproteins , Humans , Mice , Phenotype , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL