Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Nat Immunol ; 24(4): 714-728, 2023 04.
Article in English | MEDLINE | ID: mdl-36928414

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are the main source of type I interferon (IFN-I) during viral infections. Their other functions are debated, due to a lack of tools to identify and target them in vivo without affecting pDC-like cells and transitional DCs (tDCs), which harbor overlapping phenotypes and transcriptomes but a higher efficacy for T cell activation. In the present report, we present a reporter mouse, pDC-Tom, designed through intersectional genetics based on unique Siglech and Pacsin1 coexpression in pDCs. The pDC-Tom mice specifically tagged pDCs and, on breeding with Zbtb46GFP mice, enabled transcriptomic profiling of all splenic DC types, unraveling diverging activation of pDC-like cells versus tDCs during a viral infection. The pDC-Tom mice also revealed initially similar but later divergent microanatomical relocation of splenic IFN+ versus IFN- pDCs during infection. The mouse models and specific gene modules we report here will be useful to delineate the physiological functions of pDCs versus other DC types.


Subject(s)
Dendritic Cells , Interferon Type I , Animals , Mice , Interferon Type I/metabolism , Gene Expression Profiling , Phenotype , Transcriptome
2.
Nat Immunol ; 21(9): 983-997, 2020 09.
Article in English | MEDLINE | ID: mdl-32690951

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are a major source of type I interferon (IFN-I). What other functions pDCs exert in vivo during viral infections is controversial, and more studies are needed to understand their orchestration. In the present study, we characterize in depth and link pDC activation states in animals infected by mouse cytomegalovirus by combining Ifnb1 reporter mice with flow cytometry, single-cell RNA sequencing, confocal microscopy and a cognate CD4 T cell activation assay. We show that IFN-I production and T cell activation were performed by the same pDC, but these occurred sequentially in time and in different micro-anatomical locations. In addition, we show that pDC commitment to IFN-I production was marked early on by their downregulation of leukemia inhibitory factor receptor and was promoted by cell-intrinsic tumor necrosis factor signaling. We propose a new model for how individual pDCs are endowed to exert different functions in vivo during a viral infection, in a manner tightly orchestrated in time and space.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Herpesviridae Infections/immunology , Muromegalovirus/physiology , Animals , Cells, Cultured , Interferon Type I/metabolism , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Sequence Analysis, RNA , Signal Transduction , Single-Cell Analysis , Tumor Necrosis Factor-alpha/metabolism
3.
Immunity ; 47(4): 680-696.e8, 2017 10 17.
Article in English | MEDLINE | ID: mdl-29045900

ABSTRACT

The classical model of hematopoiesis established in the mouse postulates that lymphoid cells originate from a founder population of common lymphoid progenitors. Here, using a modeling approach in humanized mice, we showed that human lymphoid development stemmed from distinct populations of CD127- and CD127+ early lymphoid progenitors (ELPs). Combining molecular analyses with in vitro and in vivo functional assays, we demonstrated that CD127- and CD127+ ELPs emerged independently from lympho-mono-dendritic progenitors, responded differently to Notch1 signals, underwent divergent modes of lineage restriction, and displayed both common and specific differentiation potentials. Whereas CD127- ELPs comprised precursors of T cells, marginal zone B cells, and natural killer (NK) and innate lymphoid cells (ILCs), CD127+ ELPs supported production of all NK cell, ILC, and B cell populations but lacked T potential. On the basis of these results, we propose a "two-family" model of human lymphoid development that differs from the prevailing model of hematopoiesis.


Subject(s)
B-Lymphocytes/metabolism , Killer Cells, Natural/metabolism , Lymphoid Progenitor Cells/metabolism , Lymphopoiesis/genetics , T-Lymphocytes/metabolism , Adolescent , Adult , Animals , B-Lymphocytes/cytology , Cell Differentiation/genetics , Cell Lineage/genetics , Cells, Cultured , Female , Gene Expression Profiling/methods , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Interleukin-7 Receptor alpha Subunit/genetics , Interleukin-7 Receptor alpha Subunit/metabolism , Killer Cells, Natural/cytology , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/transplantation , Male , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Middle Aged , Stem Cell Transplantation , T-Lymphocytes/cytology , Transplantation, Heterologous , Young Adult
4.
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
5.
Cell Mol Life Sci ; 81(1): 351, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39147987

ABSTRACT

Deciphering the initial steps of SARS-CoV-2 infection, that influence COVID-19 outcomes, is challenging because animal models do not always reproduce human biological processes and in vitro systems do not recapitulate the histoarchitecture and cellular composition of respiratory tissues. To address this, we developed an innovative ex vivo model of whole human lung infection with SARS-CoV-2, leveraging a lung transplantation technique. Through single-cell RNA-seq, we identified that alveolar and monocyte-derived macrophages (AMs and MoMacs) were initial targets of the virus. Exposure of isolated lung AMs, MoMacs, classical monocytes and non-classical monocytes (ncMos) to SARS-CoV-2 variants revealed that while all subsets responded, MoMacs produced higher levels of inflammatory cytokines than AMs, and ncMos contributed the least. A Wuhan lineage appeared to be more potent than a D614G virus, in a dose-dependent manner. Amidst the ambiguity in the literature regarding the initial SARS-CoV-2 cell target, our study reveals that AMs and MoMacs are dominant primary entry points for the virus, and suggests that their responses may conduct subsequent injury, depending on their abundance, the viral strain and dose. Interfering on virus interaction with lung macrophages should be considered in prophylactic strategies.


Subject(s)
COVID-19 , Cytokines , Lung , Macrophages, Alveolar , Macrophages , SARS-CoV-2 , Humans , COVID-19/virology , COVID-19/immunology , SARS-CoV-2/physiology , Lung/virology , Lung/immunology , Lung/pathology , Macrophages/virology , Macrophages/metabolism , Macrophages/immunology , Macrophages, Alveolar/virology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Cytokines/metabolism , Monocytes/virology , Monocytes/metabolism , Monocytes/immunology , Male , Female , Single-Cell Analysis , Middle Aged
6.
EMBO J ; 37(19)2018 10 01.
Article in English | MEDLINE | ID: mdl-30131424

ABSTRACT

Plasmacytoid dendritic cells (pDC) are the major source of type I interferons (IFN-I) during viral infections, in response to triggering of endosomal Toll-like receptors (TLRs) 7 or 9 by viral single-stranded RNA or unmethylated CpG DNA, respectively. Synthetic ligands have been used to disentangle the underlying signaling pathways. The adaptor protein AP3 is necessary to transport molecular complexes of TLRs, synthetic CpG DNA, and MyD88 into endosomal compartments allowing interferon regulatory factor 7 (IRF7) recruitment whose phosphorylation then initiates IFN-I production. High basal expression of IRF7 by pDC and its further enhancement by positive IFN-I feedback signaling appear to be necessary for robust cytokine production. In contrast, we show here that in vivo during mouse cytomegalovirus (MCMV) infection pDC produce high amounts of IFN-I downstream of the TLR9-to-MyD88-to-IRF7 signaling pathway without requiring IFN-I positive feedback, high IRF7 expression, or AP3-driven endosomal routing of TLRs. Hence, the current model of the molecular requirements for professional IFN-I production by pDC, established by using synthetic TLR ligands, does not strictly apply to a physiological viral infection.


Subject(s)
Dendritic Cells/immunology , Herpesviridae Infections/immunology , Interferon Type I/immunology , Muromegalovirus/immunology , Signal Transduction/immunology , Adaptor Protein Complex 3/genetics , Adaptor Protein Complex 3/immunology , Animals , Dendritic Cells/pathology , Endosomes/genetics , Endosomes/immunology , Herpesviridae Infections/genetics , Herpesviridae Infections/pathology , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Interferon Type I/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Signal Transduction/genetics , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/immunology , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology
7.
EMBO J ; 36(6): 761-782, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28100675

ABSTRACT

In innate immune responses, induction of type-I interferons (IFNs) prevents virus spreading while viral replication is delayed by protein synthesis inhibition. We asked how cells perform these apparently contradictory activities. Using single fibroblast monitoring by flow cytometry and mathematical modeling, we demonstrate that type-I IFN production is linked to cell's ability to enter dsRNA-activated PKR-dependent translational arrest and then overcome this inhibition by decreasing eIF2α phosphorylation through phosphatase 1c cofactor GADD34 (Ppp1r15a) expression. GADD34 expression, shown here to be dependent on the IRF3 transcription factor, is responsible for a biochemical cycle permitting pulse of IFN synthesis to occur in cells undergoing protein synthesis inhibition. Translation arrest is further demonstrated to be key for anti-viral response by acting synergistically with MAVS activation to amplify TBK1 signaling and IFN-ß mRNA transcription, while GADD34-dependent protein synthesis recovery contributes to the heterogeneous expression of IFN observed in dsRNA-activated cells.


Subject(s)
Gene Expression Regulation , Interferon-beta/metabolism , Protein Biosynthesis , Protein Phosphatase 1/metabolism , RNA, Double-Stranded/immunology , RNA, Double-Stranded/metabolism , Animals , Cells, Cultured , Fibroblasts/immunology , Fibroblasts/virology , Flow Cytometry , Gene Expression Profiling , Immunity, Innate , Mice , Models, Theoretical
8.
Proc Natl Acad Sci U S A ; 114(4): E540-E549, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28069953

ABSTRACT

Immune response against pathogens is a tightly regulated process that must ensure microbial control while preserving integrity of the infected organs. Tuberculosis (TB) is a paramount example of a chronic infection in which antimicrobial immunity is protective in the vast majority of infected individuals but can become detrimental if not finely tuned. Here, we report that C-type lectin dendritic cell (DC) immunoreceptor (DCIR), a key component in DC homeostasis, is required to modulate lung inflammation and bacterial burden in TB. DCIR is abundantly expressed in pulmonary lesions in Mycobacterium tuberculosis-infected nonhuman primates during both latent and active disease. In mice, we found that DCIR deficiency impairs STAT1-mediated type I IFN signaling in DCs, leading to increased production of IL-12 and increased differentiation of T lymphocytes toward Th1 during infection. As a consequence, DCIR-deficient mice control M. tuberculosis better than WT animals but also develop more inflammation characterized by an increased production of TNF and inducible NOS (iNOS) in the lungs. Altogether, our results reveal a pathway by which a C-type lectin modulates the equilibrium between infection-driven inflammation and pathogen's control through sustaining type I IFN signaling in DCs.


Subject(s)
Dendritic Cells/immunology , Interferon Type I/immunology , Lectins, C-Type/immunology , Tuberculosis/immunology , Animals , Female , Lectins, C-Type/genetics , Macaca mulatta , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , STAT1 Transcription Factor/immunology , Signal Transduction
9.
J Immunol ; 192(10): 4510-7, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24740508

ABSTRACT

The existence of conventional dendritic cells (cDCs) has not yet been demonstrated outside mammals. In this article, we identified bona fide cDCs in chicken spleen. Comparative profiling of global and of immune response gene expression, morphology, and T cell activation properties show that cDCs and macrophages (MPs) exist as distinct mononuclear phagocytes in the chicken, resembling their human and mouse cell counterparts. With computational analysis, core gene expression signatures for cDCs, MPs, and T and B cells across the chicken, human, and mouse were established, which will facilitate the identification of these subsets in other vertebrates. Overall, this study, by extending the newly uncovered cDC and MP paradigm to the chicken, suggests that these two phagocyte lineages were already in place in the common ancestor of reptiles (including birds) and mammals in evolution. It opens avenues for the design of new vaccines and nutraceuticals that are mandatory for the sustained supply of poultry products in the expanding human population.


Subject(s)
Dendritic Cells/immunology , Gene Expression Profiling , Gene Expression Regulation/immunology , Animals , Cell Line , Chickens , Dendritic Cells/cytology , Humans , Macrophages/cytology , Macrophages/immunology , Mice , T-Lymphocytes/cytology , T-Lymphocytes/immunology
10.
J Immunol ; 193(12): 5883-93, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25385823

ABSTRACT

Swine skin is one of the best structural models for human skin, widely used to probe drug transcutaneous passage and to test new skin vaccination devices. However, little is known about its composition in immune cells, and among them dendritic cells (DC), that are essential in the initiation of the immune response. After a first seminal work describing four different DC subpopulations in pig skin, we hereafter deepen the characterization of these cells, showing the similarities between swine DC subsets and their human counterparts. Using comparative transcriptomic study, classical phenotyping as well as in vivo and in vitro functional studies, we show that swine CD163(pos) dermal DC (DDC) are transcriptomically similar to the human CD14(pos) DDC. CD163(pos) DDC are recruited in inflamed skin, they migrate in inflamed lymph but they are not attracted toward CCL21, and they modestly activate allogeneic CD8 T cells. We also show that CD163(low) DDC are transcriptomically similar to the human CD1a(pos) DDC. CD163(low) DDC migrate toward CCL21, they activate allogeneic CD8 and CD4 T cells and, like their potential human lung counterpart, they skew CD4 T cells toward a Th17 profile. We thus conclude that swine skin is a relevant model for human skin vaccination.


Subject(s)
Chemotaxis/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Langerhans Cells/immunology , Langerhans Cells/metabolism , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , Transcriptome , Animals , Antigens, CD1/genetics , Antigens, CD1/metabolism , Antigens, Surface/metabolism , Chemotaxis/genetics , Cytokines/biosynthesis , Gene Expression Profiling , Humans , Immunophenotyping , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Macrophages/immunology , Macrophages/metabolism , Mice , Phenotype , Skin/immunology , Swine
11.
J Immunol ; 193(4): 1622-35, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25009205

ABSTRACT

Human monocyte-derived dendritic cell (MoDC) have been used in the clinic with moderately encouraging results. Mouse XCR1(+) DC excel at cross-presentation, can be targeted in vivo to induce protective immunity, and share characteristics with XCR1(+) human DC. Assessment of the immunoactivation potential of XCR1(+) human DC is hindered by their paucity in vivo and by their lack of a well-defined in vitro counterpart. We report in this study a protocol generating both XCR1(+) and XCR1(-) human DC in CD34(+) progenitor cultures (CD34-DC). Gene expression profiling, phenotypic characterization, and functional studies demonstrated that XCR1(-) CD34-DC are similar to canonical MoDC, whereas XCR1(+) CD34-DC resemble XCR1(+) blood DC (bDC). XCR1(+) DC were strongly activated by polyinosinic-polycytidylic acid but not LPS, and conversely for MoDC. XCR1(+) DC and MoDC expressed strikingly different patterns of molecules involved in inflammation and in cross-talk with NK or T cells. XCR1(+) CD34-DC but not MoDC efficiently cross-presented a cell-associated Ag upon stimulation by polyinosinic-polycytidylic acid or R848, likewise to what was reported for XCR1(+) bDC. Hence, it is feasible to generate high numbers of bona fide XCR1(+) human DC in vitro as a model to decipher the functions of XCR1(+) bDC and as a potential source of XCR1(+) DC for clinical use.


Subject(s)
Antigens, CD34/immunology , Blood Cells/immunology , Dendritic Cells/immunology , Monocytes/immunology , Receptors, G-Protein-Coupled/immunology , Adjuvants, Immunologic/pharmacology , Antigen Presentation/immunology , Cell Culture Techniques , Cell Differentiation/immunology , Cell Line , Cross-Priming/immunology , Gene Expression Profiling , Green Fluorescent Proteins , Humans , Imidazoles/immunology , Killer Cells, Natural/immunology , Lipopolysaccharides/immunology , Phenotype , Poly I-C/immunology , T-Lymphocytes/immunology , Toll-Like Receptor 3 , Toll-Like Receptor 4
12.
BMC Genomics ; 16: 814, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26481321

ABSTRACT

BACKGROUND: Recent advances in the analysis of high-throughput expression data have led to the development of tools that scaled-up their focus from single-gene to gene set level. For example, the popular Gene Set Enrichment Analysis (GSEA) algorithm can detect moderate but coordinated expression changes of groups of presumably related genes between pairs of experimental conditions. This considerably improves extraction of information from high-throughput gene expression data. However, although many gene sets covering a large panel of biological fields are available in public databases, the ability to generate home-made gene sets relevant to one's biological question is crucial but remains a substantial challenge to most biologists lacking statistic or bioinformatic expertise. This is all the more the case when attempting to define a gene set specific of one condition compared to many other ones. Thus, there is a crucial need for an easy-to-use software for generation of relevant home-made gene sets from complex datasets, their use in GSEA, and the correction of the results when applied to multiple comparisons of many experimental conditions. RESULT: We developed BubbleGUM (GSEA Unlimited Map), a tool that allows to automatically extract molecular signatures from transcriptomic data and perform exhaustive GSEA with multiple testing correction. One original feature of BubbleGUM notably resides in its capacity to integrate and compare numerous GSEA results into an easy-to-grasp graphical representation. We applied our method to generate transcriptomic fingerprints for murine cell types and to assess their enrichments in human cell types. This analysis allowed us to confirm homologies between mouse and human immunocytes. CONCLUSIONS: BubbleGUM is an open-source software that allows to automatically generate molecular signatures out of complex expression datasets and to assess directly their enrichment by GSEA on independent datasets. Enrichments are displayed in a graphical output that helps interpreting the results. This innovative methodology has recently been used to answer important questions in functional genomics, such as the degree of similarities between microarray datasets from different laboratories or with different experimental models or clinical cohorts. BubbleGUM is executable through an intuitive interface so that both bioinformaticians and biologists can use it. It is available at http://www.ciml.univ-mrs.fr/applications/BubbleGUM/index.html .


Subject(s)
Computational Biology , Software , Transcriptome/genetics , Algorithms , Animals , Databases, Genetic , Gene Expression Profiling , Gene Expression Regulation , Humans , Mice , Oligonucleotide Array Sequence Analysis
13.
EMBO J ; 30(14): 2934-47, 2011 Jun 17.
Article in English | MEDLINE | ID: mdl-21685873

ABSTRACT

The gut is a major barrier against microbes and encloses various innate lymphoid cells (ILCs), including two subsets expressing the natural cytotoxicity receptor NKp46. A subset of NKp46(+) cells expresses retinoic acid receptor-related orphan receptor γt (RORγt) and produces IL-22, like lymphoid tissue inducer (LTi) cells. Other NKp46(+) cells lack RORγt and produce IFN-γ, like conventional Natural Killer (cNK) cells. The identity, the regulation and the in vivo functions of gut NKp46(+) ILCs largely remain to be unravelled. Using pan-genomic profiling, we showed here that small intestine (SI) NKp46(+)RORγt(-) ILCs correspond to SI NK cells. Conversely, we identified a transcriptional programme conserved in fetal LTi cells and adult SI NKp46(+)RORγt(+) and NKp46(-)RORγt(+) ILCs. We also demonstrated that the IL-1ß/IL-1R1/MyD88 pathway, but not the commensal flora, drove IL-22 production by NKp46(+)RORγt(+) ILCs. Finally, oral Listeria monocytogenes infection induced IFN-γ production in SI NK and IL-22 production in NKp46(+)RORγt(+) ILCs, but only IFN-γ contributed to control bacteria dissemination. NKp46(+) ILC heterogeneity is thus associated with subset-specific transcriptional programmes and effector functions that govern their implication in gut innate immunity.


Subject(s)
Cell Lineage , Immunity, Innate , Lymphocytes/metabolism , Lymphocytes/microbiology , Natural Cytotoxicity Triggering Receptor 1/metabolism , Receptors, Retinoic Acid/metabolism , Animals , Female , Flow Cytometry , Intestine, Small/immunology , Intestine, Small/metabolism , Intestine, Small/microbiology , Listeria monocytogenes/isolation & purification , Listeriosis/metabolism , Listeriosis/microbiology , Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/physiology , Natural Cytotoxicity Triggering Receptor 1/genetics , Receptors, Interleukin-1/physiology , Receptors, Retinoic Acid/genetics , Tissue Distribution , Retinoic Acid Receptor gamma
14.
Proc Natl Acad Sci U S A ; 108(41): 17016-21, 2011 Oct 11.
Article in English | MEDLINE | ID: mdl-21969581

ABSTRACT

Earlier work has shown that pre-B cells can be converted into macrophages by the transcription factor CCAAT/enhancer binding protein α at very high frequencies. Using this system, we performed a systematic analysis of whether during transdifferentiation the cells transiently reactivate progenitor-restricted genes or even retrodifferentiate. A transcriptome analysis of transdifferentiating cells showed that most genes are up- or down-regulated continuously, acquiring a macrophage phenotype within 5 d. In addition, we observed the transient reactivation of a subset of immature myeloid markers, as well as low levels of the progenitor markers Kit and FMS-like tyrosine kinase 3 and a few lineage-inappropriate genes. Importantly, however, we were unable to observe the reexpression of cell-surface marker combinations that characterize hematopoietic stem and progenitor cells, including c-Kit and FMS-like tyrosine kinase 3, even when CAAT/enhancer binding protein α was activated in pre-B cells under culture conditions that favor growth of hematopoietic stem and progenitor cells or when the transcription factor was activated in a time-limited fashion. Together, our findings are consistent with the notion that the conversion from pre-B cells to macrophages is mostly direct and does not involve overt retrodifferentiation.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/physiology , Cell Transdifferentiation/physiology , Macrophages/cytology , Macrophages/metabolism , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/metabolism , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , Cell Lineage/genetics , Cell Transdifferentiation/genetics , Cells, Cultured , Erythrocytes/cytology , Erythrocytes/metabolism , Genes, cdc , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice , Proto-Oncogene Proteins c-kit/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Transcriptome , fms-Like Tyrosine Kinase 3/genetics
15.
Elife ; 122024 Jun 26.
Article in English | MEDLINE | ID: mdl-38922679

ABSTRACT

During tuberculosis (TB), migration of dendritic cells (DCs) from the site of infection to the draining lymph nodes is known to be impaired, hindering the rapid development of protective T-cell-mediated immunity. However, the mechanisms involved in the delayed migration of DCs during TB are still poorly defined. Here, we found that infection of DCs with Mycobacterium tuberculosis (Mtb) triggers HIF1A-mediated aerobic glycolysis in a TLR2-dependent manner, and that this metabolic profile is essential for DC migration. In particular, the lactate dehydrogenase inhibitor oxamate and the HIF1A inhibitor PX-478 abrogated Mtb-induced DC migration in vitro to the lymphoid tissue-specific chemokine CCL21, and in vivo to lymph nodes in mice. Strikingly, we found that although monocytes from TB patients are inherently biased toward glycolysis metabolism, they differentiate into poorly glycolytic and poorly migratory DCs compared with healthy subjects. Taken together, these data suggest that because of their preexisting glycolytic state, circulating monocytes from TB patients are refractory to differentiation into migratory DCs, which may explain the delayed migration of these cells during the disease and opens avenues for host-directed therapies for TB.


Subject(s)
Cell Movement , Dendritic Cells , Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit , Monocytes , Mycobacterium tuberculosis , Tuberculosis , Dendritic Cells/metabolism , Dendritic Cells/immunology , Monocytes/metabolism , Monocytes/immunology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mycobacterium tuberculosis/immunology , Animals , Tuberculosis/immunology , Tuberculosis/metabolism , Tuberculosis/microbiology , Mice , Toll-Like Receptor 2/metabolism , Mice, Inbred C57BL , Female
16.
J Immunol ; 187(9): 4411-5, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21948982

ABSTRACT

Subsets of dendritic cells (DCs) have been described according to their functions and anatomical locations. Conventional DC subsets are defined by reciprocal expression of CD11b and CD8α in lymphoid tissues (LT), and of CD11b and CD103 in non-LT (NLT). Spleen CD8α(+) and dermal CD103(+) DCs share a high efficiency for Ag cross-presentation and a developmental dependency on specific transcription factors. However, it is not known whether all NLT-derived CD103(+) DCs and LT-resident CD8α(+) DCs are similar despite their different anatomical locations. XCR1 was previously described as exclusively expressed on mouse spleen CD8α(+) DCs and human blood BDCA3(+) DCs. In this article, we showed that LT-resident CD8α(+) DCs and NLT-derived CD103(+) DCs specifically express XCR1 and are characterized by a unique transcriptional fingerprint, irrespective of their tissue of origin. Therefore, CD8α(+) DCs and CD103(+) DCs belong to a common DC subset which is unequivocally identified by XCR1 expression throughout the body.


Subject(s)
CD8 Antigens/biosynthesis , Cell Movement/immunology , Dendritic Cells/immunology , Lymphoid Tissue/immunology , Receptors, Chemokine/biosynthesis , Receptors, G-Protein-Coupled/biosynthesis , Animals , Antigens, CD/biosynthesis , Cell Movement/genetics , DNA Fingerprinting , Dendritic Cells/classification , Dendritic Cells/cytology , Genetic Markers/immunology , Humans , Integrin alpha Chains/biosynthesis , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Chemokine/genetics , Transcription, Genetic/immunology
17.
Methods Mol Biol ; 2618: 319-373, 2023.
Article in English | MEDLINE | ID: mdl-36905526

ABSTRACT

Dendritic cells (DCs) orchestrate innate and adaptive immunity, by translating the sensing of distinct danger signals into the induction of different effector lymphocyte responses, to induce the defense mechanisms the best suited to face the threat. Hence, DCs are very plastic, which results from two key characteristics. First, DCs encompass distinct cell types specialized in different functions. Second, each DC type can undergo different activation states, fine-tuning its functions depending on its tissue microenvironment and the pathophysiological context, by adapting the output signals it delivers to the input signals it receives. Hence, to better understand DC biology and harness it in the clinic, we must determine which combinations of DC types and activation states mediate which functions and how.To decipher the nature, functions, and regulation of DC types and their physiological activation states, one of the methods that can be harnessed most successfully is ex vivo single-cell RNA sequencing (scRNAseq). However, for new users of this approach, determining which analytics strategy and computational tools to choose can be quite challenging, considering the rapid evolution and broad burgeoning in the field. In addition, awareness must be raised on the need for specific, robust, and tractable strategies to annotate cells for cell type identity and activation states. It is also important to emphasize the necessity of examining whether similar cell activation trajectories are inferred by using different, complementary methods. In this chapter, we take these issues into account for providing a pipeline for scRNAseq analysis and illustrating it with a tutorial reanalyzing a public dataset of mononuclear phagocytes isolated from the lungs of naïve or tumor-bearing mice. We describe this pipeline step-by-step, including data quality controls, dimensionality reduction, cell clustering, cell cluster annotation, inference of the cell activation trajectories, and investigation of the underpinning molecular regulation. It is accompanied with a more complete tutorial on GitHub. We hope that this method will be helpful for both wet lab and bioinformatics researchers interested in harnessing scRNAseq data for deciphering the biology of DCs or other cell types and that it will contribute to establishing high standards in the field.


Subject(s)
Dendritic Cells , Neoplasms , Animals , Mice , Computational Biology , Neoplasms/metabolism , Sequence Analysis, RNA , Tumor Microenvironment
18.
Front Immunol ; 14: 1142228, 2023.
Article in English | MEDLINE | ID: mdl-37465668

ABSTRACT

In response to the increasing demand for lung transplantation, ex vivo lung perfusion (EVLP) has extended the number of suitable donor lungs by rehabilitating marginal organs. However despite an expanding use in clinical practice, the responses of the different lung cell types to EVLP are not known. In order to advance our mechanistic understanding and establish a refine tool for improvement of EVLP, we conducted a pioneer study involving single cell RNA-seq on human lungs declined for transplantation. Functional enrichment analyses were performed upon integration of data sets generated at 4 h (clinical duration) and 10 h (prolonged duration) from two human lungs processed to EVLP. Pathways related to inflammation were predicted activated in epithelial and blood endothelial cells, in monocyte-derived macrophages and temporally at 4 h in alveolar macrophages. Pathways related to cytoskeleton signaling/organization were predicted reduced in most cell types mainly at 10 h. We identified a division of labor between cell types for the selected expression of cytokine and chemokine genes that varied according to time. Immune cells including CD4+ and CD8+ T cells, NK cells, mast cells and conventional dendritic cells displayed gene expression patterns indicating blunted activation, already at 4 h in several instances and further more at 10 h. Therefore despite inducing inflammatory responses, EVLP appears to dampen the activation of major lung immune cell types, what may be beneficial to the outcome of transplantation. Our results also support that therapeutics approaches aiming at reducing inflammation upon EVLP should target both the alveolar and vascular compartments.


Subject(s)
CD8-Positive T-Lymphocytes , Lung Transplantation , Humans , Perfusion/methods , Endothelial Cells , Lung Transplantation/methods , Lung/physiology , Inflammation
19.
J Immunol ; 185(6): 3313-25, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20702727

ABSTRACT

The mouse lymphoid organ-resident CD8alpha(+) dendritic cell (DC) subset is specialized in Ag presentation to CD8(+) T cells. Recent evidence shows that mouse nonlymphoid tissue CD103(+) DCs and human blood DC Ag 3(+) DCs share similarities with CD8alpha(+) DCs. We address here whether the organization of DC subsets is conserved across mammals in terms of gene expression signatures, phenotypic characteristics, and functional specialization, independently of the tissue of origin. We study the DC subsets that migrate from the skin in the ovine species that, like all domestic animals, belongs to the Laurasiatheria, a distinct phylogenetic clade from the supraprimates (human/mouse). We demonstrate that the minor sheep CD26(+) skin lymph DC subset shares significant transcriptomic similarities with mouse CD8alpha(+) and human blood DC Ag 3(+) DCs. This allowed the identification of a common set of phenotypic characteristics for CD8alpha-like DCs in the three mammalian species (i.e., SIRP(lo), CADM1(hi), CLEC9A(hi), CD205(hi), XCR1(hi)). Compared to CD26(-) DCs, the sheep CD26(+) DCs show 1) potent stimulation of allogeneic naive CD8(+) T cells with high selective induction of the Ifngamma and Il22 genes; 2) dominant efficacy in activating specific CD8(+) T cells against exogenous soluble Ag; and 3) selective expression of functional pathways associated with high capacity for Ag cross-presentation. Our results unravel a unifying definition of the CD8alpha(+)-like DCs across mammalian species and identify molecular candidates that could be used for the design of vaccines applying to mammals in general.


Subject(s)
CD8 Antigens/biosynthesis , Conserved Sequence , Dendritic Cells/immunology , Gene Expression Profiling/methods , Lymph/cytology , Lymph/immunology , Animals , CD8 Antigens/physiology , Cells, Cultured , Dendritic Cells/metabolism , Dipeptidyl Peptidase 4/biosynthesis , Female , Humans , Immune Tolerance , Lymph/metabolism , Mice , Sheep, Domestic , Skin/cytology , Skin/immunology , Skin/metabolism , Species Specificity
20.
Front Immunol ; 11: 588411, 2020.
Article in English | MEDLINE | ID: mdl-33365028

ABSTRACT

Porcine reproductive and respiratory syndrome (PRRS) has an extensive impact on pig production. The causative virus (PRRSV) is divided into two species, PRRSV-1 (European origin) and PRRSV-2 (North American origin). Within PRRSV-1, PRRSV-1.3 strains, such as Lena, are more pathogenic than PRRSV-1.1 strains, such as Flanders 13 (FL13). To date, the molecular interactions of PRRSV with primary lung mononuclear phagocyte (MNP) subtypes, including conventional dendritic cells types 1 (cDC1) and 2 (cDC2), monocyte-derived DCs (moDC), and pulmonary intravascular macrophages (PIM), have not been thoroughly investigated. Here, we analyze the transcriptome profiles of in vivo FL13-infected parenchymal MNP subpopulations and of in vitro FL13- and Lena-infected parenchymal MNP. The cell-specific expression profiles of in vivo sorted cells correlated with their murine counterparts (AM, cDC1, cDC2, moDC) with the exception of PIM. Both in vivo and in vitro, FL13 infection altered the expression of a low number of host genes, and in vitro infection with Lena confirmed the higher ability of this strain to modulate host response. Machine learning (ML) and gene set enrichment analysis (GSEA) unraveled additional relevant genes and pathways modulated by FL13 infection that were not identified by conventional analyses. GSEA increased the cellular pathways enriched in the FL13 data set, but ML allowed a more complete comprehension of functional profiles during FL13 in vitro infection. Data indicates that cellular reprogramming differs upon Lena and FL13 infection and that the latter might keep antiviral and inflammatory macrophage/DC functions silent. Although the slow replication kinetics of FL13 likely contribute to differences in cellular gene expression, the data suggest distinct mechanisms of interaction of the two viruses with the innate immune system during early infection.


Subject(s)
Monocytes/immunology , Porcine Reproductive and Respiratory Syndrome/genetics , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine respiratory and reproductive syndrome virus , Animals , Female , Lung/cytology , Monocytes/virology , Swine , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL