Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Cell ; 187(4): 962-980.e19, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38309258

ABSTRACT

Microglia (MG), the brain-resident macrophages, play major roles in health and disease via a diversity of cellular states. While embryonic MG display a large heterogeneity of cellular distribution and transcriptomic states, their functions remain poorly characterized. Here, we uncovered a role for MG in the maintenance of structural integrity at two fetal cortical boundaries. At these boundaries between structures that grow in distinct directions, embryonic MG accumulate, display a state resembling post-natal axon-tract-associated microglia (ATM) and prevent the progression of microcavities into large cavitary lesions, in part via a mechanism involving the ATM-factor Spp1. MG and Spp1 furthermore contribute to the rapid repair of lesions, collectively highlighting protective functions that preserve the fetal brain from physiological morphogenetic stress and injury. Our study thus highlights key major roles for embryonic MG and Spp1 in maintaining structural integrity during morphogenesis, with major implications for our understanding of MG functions and brain development.


Subject(s)
Brain , Microglia , Axons , Brain/cytology , Brain/growth & development , Macrophages/physiology , Microglia/pathology , Morphogenesis
2.
Cell ; 185(7): 1189-1207.e25, 2022 03 31.
Article in English | MEDLINE | ID: mdl-35325594

ABSTRACT

Macrophage infiltration is a hallmark of solid cancers, and overall macrophage infiltration correlates with lower patient survival and resistance to therapy. Tumor-associated macrophages, however, are phenotypically and functionally heterogeneous. Specific subsets of tumor-associated macrophage might be endowed with distinct roles on cancer progression and antitumor immunity. Here, we identify a discrete population of FOLR2+ tissue-resident macrophages in healthy mammary gland and breast cancer primary tumors. FOLR2+ macrophages localize in perivascular areas in the tumor stroma, where they interact with CD8+ T cells. FOLR2+ macrophages efficiently prime effector CD8+ T cells ex vivo. The density of FOLR2+ macrophages in tumors positively correlates with better patient survival. This study highlights specific roles for tumor-associated macrophage subsets and paves the way for subset-targeted therapeutic interventions in macrophages-based cancer therapies.


Subject(s)
Breast Neoplasms , Macrophages , Breast/immunology , Breast Neoplasms/epidemiology , Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes , Female , Folate Receptor 2 , Humans , Lymphocytes, Tumor-Infiltrating , Prognosis
3.
Annu Rev Immunol ; 31: 563-604, 2013.
Article in English | MEDLINE | ID: mdl-23516985

ABSTRACT

Dendritic cells (DCs) form a remarkable cellular network that shapes adaptive immune responses according to peripheral cues. After four decades of research, we now know that DCs arise from a hematopoietic lineage distinct from other leukocytes, establishing the DC system as a unique hematopoietic branch. Recent work has also established that tissue DCs consist of developmentally and functionally distinct subsets that differentially regulate T lymphocyte function. This review discusses major advances in our understanding of the regulation of DC lineage commitment, differentiation, diversification, and function in situ.


Subject(s)
Cell Differentiation/immunology , Cell Lineage/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Animals , Cell Movement/immunology , Dendritic Cells/cytology , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/physiology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Lymphoid Tissue/pathology
4.
Immunity ; 55(1): 129-144.e8, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34910930

ABSTRACT

Dendritic cells (DCs) patrol tissues and transport antigens to lymph nodes to initiate adaptive immune responses. Within tissues, DCs constitute a complex cell population composed of distinct subsets that can exhibit different activation states and functions. How tissue-specific cues orchestrate DC diversification remains elusive. Here, we show that the small intestine included two pools of cDC2s originating from common pre-DC precursors: (1) lamina propria (LP) CD103+CD11b+ cDC2s that were mature-like proinflammatory cells and (2) intraepithelial cDC2s that exhibited an immature-like phenotype as well as tolerogenic properties. These phenotypes resulted from the action of food-derived retinoic acid (ATRA), which enhanced actomyosin contractility and promoted LP cDC2 transmigration into the epithelium. There, cDC2s were imprinted by environmental cues, including ATRA itself and the mucus component Muc2. Hence, by reaching distinct subtissular niches, DCs can exist as immature and mature cells within the same tissue, revealing an additional mechanism of DC functional diversification.


Subject(s)
Dendritic Cells/immunology , Inflammation/immunology , Intestinal Mucosa/pathology , T-Lymphocytes/immunology , Actomyosin/metabolism , Animals , Antigen Presentation , Antigens, CD/metabolism , CD11b Antigen/metabolism , Cell Differentiation , Cell Movement , Cells, Cultured , Immune Tolerance , Integrin alpha Chains/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucin-2/immunology , Tretinoin/metabolism
5.
Immunity ; 53(2): 335-352.e8, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32610077

ABSTRACT

Dendritic cells (DCs) are antigen-presenting cells controlling T cell activation. In humans, the diversity, ontogeny, and functional capabilities of DC subsets are not fully understood. Here, we identified circulating CD88-CD1c+CD163+ DCs (called DC3s) as immediate precursors of inflammatory CD88-CD14+CD1c+CD163+FcεRI+ DCs. DC3s develop via a specific pathway activated by GM-CSF, independent of cDC-restricted (CDP) and monocyte-restricted (cMoP) progenitors. Like classical DCs but unlike monocytes, DC3s drove activation of naive T cells. In vitro, DC3s displayed a distinctive ability to prime CD8+ T cells expressing a tissue homing signature and the epithelial homing alpha-E integrin (CD103) through transforming growth factor ß (TGF-ß) signaling. In vivo, DC3s infiltrated luminal breast cancer primary tumors, and DC3 infiltration correlated positively with CD8+CD103+CD69+ tissue-resident memory T cells. Together, these findings define DC3s as a lineage of inflammatory DCs endowed with a strong potential to regulate tumor immunity.


Subject(s)
Antigens, CD1/metabolism , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes/cytology , Dendritic Cells/immunology , Glycoproteins/metabolism , Integrin alpha Chains/metabolism , Receptors, Cell Surface/metabolism , Animals , CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Line, Tumor , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD , Transforming Growth Factor beta1/metabolism , fms-Like Tyrosine Kinase 3/metabolism
7.
Nat Immunol ; 13(11): 1118-28, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23023392

ABSTRACT

We assessed gene expression in tissue macrophages from various mouse organs. The diversity in gene expression among different populations of macrophages was considerable. Only a few hundred mRNA transcripts were selectively expressed by macrophages rather than dendritic cells, and many of these were not present in all macrophages. Nonetheless, well-characterized surface markers, including MerTK and FcγR1 (CD64), along with a cluster of previously unidentified transcripts, were distinctly and universally associated with mature tissue macrophages. TCEF3, C/EBP-α, Bach1 and CREG-1 were among the transcriptional regulators predicted to regulate these core macrophage-associated genes. The mRNA encoding other transcription factors, such as Gata6, was associated with single macrophage populations. We further identified how these transcripts and the proteins they encode facilitated distinguishing macrophages from dendritic cells.


Subject(s)
Antigens, CD/genetics , Macrophages/metabolism , RNA, Messenger/genetics , Transcription Factors/genetics , Transcription, Genetic , Animals , Antigens, CD/immunology , Cell Differentiation , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation , Genetic Variation , Liver/cytology , Liver/immunology , Liver/metabolism , Lung/cytology , Lung/immunology , Lung/metabolism , Macrophages/cytology , Macrophages/immunology , Mice , Microglia/cytology , Microglia/immunology , Microglia/metabolism , Oligonucleotide Array Sequence Analysis , Organ Specificity , RNA, Messenger/immunology , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Transcription Factors/immunology
8.
Nat Immunol ; 13(9): 888-99, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22797772

ABSTRACT

Although much progress has been made in the understanding of the ontogeny and function of dendritic cells (DCs), the transcriptional regulation of the lineage commitment and functional specialization of DCs in vivo remains poorly understood. We made a comprehensive comparative analysis of CD8(+), CD103(+), CD11b(+) and plasmacytoid DC subsets, as well as macrophage DC precursors and common DC precursors, across the entire immune system. Here we characterized candidate transcriptional activators involved in the commitment of myeloid progenitor cells to the DC lineage and predicted regulators of DC functional diversity in tissues. We identified a molecular signature that distinguished tissue DCs from macrophages. We also identified a transcriptional program expressed specifically during the steady-state migration of tissue DCs to the draining lymph nodes that may control tolerance to self tissue antigens.


Subject(s)
Cell Differentiation/immunology , Cell Lineage/immunology , Dendritic Cells/immunology , Transcription, Genetic , Cell Differentiation/genetics , Dendritic Cells/cytology , Gene Expression Profiling , Humans
9.
Immunity ; 42(6): 1197-211, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26084029

ABSTRACT

Dendritic cells (DCs) are key players in the immune system. Much of their biology has been elucidated via culture systems in which hematopoietic precursors differentiate into DCs under the aegis of cytokines. A widely used protocol involves the culture of murine bone marrow (BM) cells with granulocyte-macrophage colony-stimulating factor (GM-CSF) to generate BM-derived DCs (BMDCs). BMDCs express CD11c and MHC class II (MHCII) molecules and share with DCs isolated from tissues the ability to present exogenous antigens to T cells and to respond to microbial stimuli by undergoing maturation. We demonstrate that CD11c(+)MHCII(+) BMDCs are in fact a heterogeneous group of cells that comprises conventional DCs and monocyte-derived macrophages. DCs and macrophages in GM-CSF cultures both undergo maturation upon stimulation with lipopolysaccharide but respond differentially to the stimulus and remain separable entities. These results have important implications for the interpretation of a vast array of data obtained with DC culture systems.


Subject(s)
Bone Marrow Cells/immunology , Dendritic Cells/immunology , Macrophages/immunology , Animals , Antigen Presentation , CD11c Antigen/metabolism , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Histocompatibility Antigens Class II/metabolism , Immunophenotyping , Lipopolysaccharides/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Transcriptome
10.
Immunity ; 38(4): 818-30, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23562160

ABSTRACT

In comparison to murine dendritic cells (DCs), less is known about the function of human DCs in tissues. Here, we analyzed, by using lung tissues from humans and humanized mice, the role of human CD1c(+) and CD141(+) DCs in determining the type of CD8(+) T cell immunity generated to live-attenuated influenza virus (LAIV) vaccine. We found that both lung DC subsets acquired influenza antigens in vivo and expanded specific cytotoxic CD8(+) T cells in vitro. However, lung-tissue-resident CD1c(+) DCs, but not CD141(+) DCs, were able to drive CD103 expression on CD8(+) T cells and promoted CD8(+) T cell accumulation in lung epithelia in vitro and in vivo. CD1c(+) DCs induction of CD103 expression was dependent on membrane-bound cytokine TGF-ß1. Thus, CD1c(+) and CD141(+) DCs generate CD8(+) T cells with different properties, and CD1c(+) DCs specialize in the regulation of mucosal CD8(+) T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Lung/immunology , T-Lymphocyte Subsets/immunology , Transforming Growth Factor beta/metabolism , Animals , Antigens, CD/metabolism , Antigens, CD1/metabolism , Antigens, Viral/immunology , Cell Differentiation , Cells, Cultured , Cytotoxicity, Immunologic , Glycoproteins/metabolism , Humans , Immunity, Mucosal , Immunologic Memory , Influenza Vaccines/immunology , Integrin alpha Chains/metabolism , Lung/virology , Lymphocyte Activation , Mice , Mice, SCID , Microarray Analysis
11.
Immunity ; 36(6): 1031-46, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22749353

ABSTRACT

GM-CSF (Csf-2) is a critical cytokine for the in vitro generation of dendritic cells (DCs) and is thought to control the development of inflammatory DCs and resident CD103(+) DCs in some tissues. Here we showed that in contrast to the current understanding, Csf-2 receptor acts in the steady state to promote the survival and homeostasis of nonlymphoid tissue-resident CD103(+) and CD11b(+) DCs. Absence of Csf-2 receptor on lung DCs abrogated the induction of CD8(+) T cell immunity after immunization with particulate antigens. In contrast, Csf-2 receptor was dispensable for the differentiation and innate function of inflammatory DCs during acute injuries. Instead, inflammatory DCs required Csf-1 receptor for their development. Thus, Csf-2 is important in vaccine-induced CD8(+) T cell immunity through the regulation of nonlymphoid tissue DC homeostasis rather than control of inflammatory DCs in vivo.


Subject(s)
Cytokine Receptor Common beta Subunit/physiology , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Inflammation/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Cell Lineage , Cytokine Receptor Common beta Subunit/antagonists & inhibitors , Cytokine Receptor Common beta Subunit/deficiency , Cytokine Receptor Common beta Subunit/genetics , Dendritic Cells/classification , Dendritic Cells/cytology , Encephalomyelitis, Autoimmune, Experimental/immunology , Endotoxemia/immunology , Gene Expression Profiling , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Homeostasis , Lipopolysaccharides/toxicity , Listeriosis/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/transplantation , Organ Specificity , Orthomyxoviridae Infections/immunology , Pneumococcal Infections/immunology , Radiation Chimera , Spleen/immunology , Tamoxifen/pharmacology
13.
Immunity ; 31(2): 232-44, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19699172

ABSTRACT

Dendritic cells (DCs) have the striking ability to cross-present exogenous antigens in association with major histocompatibility complex (MHC) class I to CD8(+) T cells. However, the intracellular pathways underlying cross-presentation remain ill defined. Current models involve cytosolic proteolysis of antigens by the proteasome and peptide import into endoplasmic reticulum (ER) or phagosomal lumen by the transporters associated with antigen processing (TAP1 and TAP2). Here, we show that DCs expressed an ER-resident 47 kDa immune-related GTPase, Igtp (Irgm3). Igtp resides on ER and lipid body (LB) membranes where it binds the LB coat component ADFP. Inactivation of genes encoding for either Igtp or ADFP led to defects in LB formation in DCs and severely impaired cross-presentation of phagocytosed antigens to CD8(+) T cells but not antigen presentation to CD4(+) T cells. We thus define a new role for LB organelles in regulating cross-presentation of exogenous antigens to CD8(+) T lymphocytes in DCs.


Subject(s)
Antigen Presentation/immunology , Cross-Priming , Dendritic Cells/immunology , Histocompatibility Antigens Class I/immunology , Lipids/immunology , Phagocytosis , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Endoplasmic Reticulum/immunology , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/immunology , GTP Phosphohydrolases/metabolism , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Perilipin-2
14.
Immunity ; 31(3): 513-25, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19733489

ABSTRACT

CX(3)CR1(+) and CD103(+) dendritic cells (DCs) in intestinal lamina propria play a key role in mucosal immunity. However, the origin and the developmental pathways that regulate their differentiation in the lamina propria remain unclear. We showed that monocytes gave rise exclusively to CD103(-)CX(3)CR1(+) lamina propria DCs under the control of macrophage-colony-stimulating factor receptor (M-CSFR) and Fms-like thyrosine kinase 3 (Flt3) ligands. In contrast, common DC progenitors (CDP) and pre-DCs, which give rise to lymphoid organ DCs but not to monocytes, differentiated exclusively into CD103(+)CX(3)CR1(-) lamina propria DCs under the control of Flt3 and granulocyte-macrophage-colony-stimulating factor receptor (GM-CSFR) ligands. CD103(+)CX(3)CR1(-) DCs but not CD103(-)CX(3)CR1(+) DCs in the lamina propria constitutively expressed CCR7 and were the first DCs to transport pathogenic Salmonella from the intestinal tract to the mesenteric lymph nodes. Altogether, these results underline the diverse origin of the lamina propria DC network and identify mucosal DCs that arise from pre-DCs as key sentinels of the gut immune system.


Subject(s)
Cell Lineage , Dendritic Cells/cytology , Dendritic Cells/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Animals , Antigens, CD/immunology , CX3C Chemokine Receptor 1 , Cell Differentiation , Cell Movement , Integrin alpha Chains/immunology , Lymph Nodes/immunology , Mice , Mice, Knockout , Phenotype , Receptor, Macrophage Colony-Stimulating Factor/immunology , Receptors, Chemokine/immunology , Salmonella/immunology , Salmonella/pathogenicity , fms-Like Tyrosine Kinase 3/deficiency , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/immunology
15.
J Immunol ; 195(10): 5066-76, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26459350

ABSTRACT

Dendritic cells (DCs) are powerful APCs that can induce Ag-specific adaptive immune responses and are increasingly recognized as important players in innate immunity to both infection and malignancy. Interestingly, although there are multiple described hematological malignancies, DC cancers are rarely observed in humans. Whether this is linked to the immunogenic potential of DCs, which might render them uniquely susceptible to immune control upon neoplastic transformation, has not been fully investigated. To address the issue, we generated a genetically engineered mouse model in which expression of Cre recombinase driven by the C-type lectin domain family 9, member a (Clec9a) locus causes expression of the Kirsten rat sarcoma viral oncogene homolog (Kras)(G12D) oncogenic driver and deletion of the tumor suppressor p53 within developing and differentiated DCs. We show that these Clec9a(Kras-G12D) mice rapidly succumb from disease and display massive accumulation of transformed DCs in multiple organs. In bone marrow chimeras, the development of DC cancer could be induced by a small number of transformed cells and was not prevented by the presence of untransformed DCs. Notably, activation of transformed DCs did not happen spontaneously but could be induced upon stimulation. Although Clec9a(Kras-G12D) mice showed altered thymic T cell development, peripheral T cells were largely unaffected during DC cancer development. Interestingly, transformed DCs were rejected upon adoptive transfer into wild-type but not lymphocyte-deficient mice, indicating that immunological control of DC cancer is in principle possible but does not occur during spontaneous generation in Clec9a(Kras-G12D) mice. Our findings suggest that neoplastic transformation of DCs does not by default induce anti-cancer immunity and can develop unhindered by immunological barriers.


Subject(s)
Cell Transformation, Neoplastic/immunology , Dendritic Cells/immunology , Neoplasms, Experimental/immunology , Stem Cells/immunology , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Dendritic Cells/pathology , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Mice , Mice, Transgenic , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/immunology , Rats , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Stem Cells/pathology
16.
Blood ; 119(7): 1693-701, 2012 Feb 16.
Article in English | MEDLINE | ID: mdl-22174156

ABSTRACT

R-Ras is a member of the RAS superfamily of small GTP-binding proteins. The physiologic function of R-Ras has not been fully elucidated. We found that R-Ras is expressed by lymphoid and nonlymphoid tissues and drastically up-regulated when bone marrow progenitors are induced to differentiate into dendritic cells (DCs). To address the role of R-Ras in DC functions, we generated a R-Ras-deficient mouse strain. We found that tumors induced in Rras(-/-) mice formed with shorter latency and attained greater tumor volumes. This finding has prompted the investigation of a role for R-Ras in the immune system. Indeed, Rras(-/-) mice were impaired in their ability to prime allogeneic and antigen-specific T-cell responses. Rras(-/-) DCs expressed lower levels of surface MHC class II and CD86 in response to lipopolysaccharide compared with wild-type DCs. This was correlated with a reduced phosphorylation of p38 and Akt. Consistently, R-Ras-GTP level was increased within 10 minutes of lipopolysaccharide stimulation. Furthermore, Rras(-/-) DCs have attenuated capacity to spread on fibronectin and form stable immunologic synapses with T cells. Altogether, these findings provide the first demonstration of a role for R-Ras in cell-mediated immunity and further expand on the complexity of small G-protein signaling in DCs.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/genetics , Dendritic Cells/physiology , Lymphocyte Activation/genetics , ras Proteins/physiology , Animals , Antigen Presentation/genetics , Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/metabolism , Female , Immunity, Cellular/genetics , Immunity, Cellular/immunology , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , ras Proteins/genetics , ras Proteins/metabolism
17.
Immunol Rev ; 234(1): 55-75, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20193012

ABSTRACT

Dendritic cells (DCs) have been extensively studied in mice lymphoid organs, but less is known about the origin and the mechanisms that regulate DC development and function in non-lymphoid tissues. Here, we discuss recent evidence establishing the contribution of the DC-restricted lineage to the non-lymphoid tissue DC pool and discuss the mechanisms that control the homeostasis of non-lymphoid tissue DCs. We also review recent results underlining the functional specialization of tissue DCs and discuss the potential implications of these findings in tissue immunity and in the development of novel vaccine strategies.


Subject(s)
Cell Differentiation , Cell Lineage , Dendritic Cells/immunology , Hematopoietic Stem Cells/immunology , Animals , Cell Differentiation/immunology , Cell Lineage/immunology , Cytokines/immunology , Dendritic Cells/metabolism , Hematopoietic Stem Cells/metabolism , Homeostasis , Inflammation Mediators/immunology , Mice , Phenotype , Transcription Factors/metabolism , Vaccines/immunology
18.
Methods Mol Biol ; 2618: 83-92, 2023.
Article in English | MEDLINE | ID: mdl-36905510

ABSTRACT

Dendritic cells (DCs) are mononuclear phagocytes of hematopoietic origin residing in lymphoid and nonlymphoid tissues. DCs are often referred as the sentinels of the immune system as they can sense pathogens and danger signals. Upon activation, DCs migrate to the draining lymph nodes and present antigens to naïve T cells to trigger adaptive immunity. Hematopoietic progenitors for DCs reside in the adult bone marrow (BM). Therefore, BM cell culture systems have been developed to generate large amounts of primary DCs in vitro conveniently enabling to analyze their developmental and functional features. Here, we review various protocols enabling to generate DCs in vitro from murine BM cells and discuss the cellular heterogeneity of each culture system.


Subject(s)
Bone Marrow , T-Lymphocytes , Animals , Mice , Bone Marrow Cells , Cell Differentiation , Cells, Cultured , Dendritic Cells , Mice, Inbred C57BL
19.
Methods Mol Biol ; 2618: 121-132, 2023.
Article in English | MEDLINE | ID: mdl-36905513

ABSTRACT

Dendritic cells (DCs) are professional antigen-presenting cells controlling the activation of T cells and thus regulating adaptive immune response against pathogens or tumors. Modeling human DC differentiation and function is crucial for our understanding of immune response and the development of new therapies. Considering DC rarity in human blood, in vitro systems allowing their faithful generation are needed. This chapter will describe a DC differentiation method based on the co-culture of CD34+ cord blood progenitors together with mesenchymal stromal cells (eMSCs) engineered to deliver growth factors and chemokines.


Subject(s)
Dendritic Cells , Fetal Blood , Humans , Cells, Cultured , Antigens, CD34/metabolism , Cell Differentiation , Cell Adhesion Molecules
20.
Nat Cell Biol ; 25(12): 1736-1745, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38036749

ABSTRACT

Myeloid cell infiltration of solid tumours generally associates with poor patient prognosis and disease severity1-13. Therefore, understanding the regulation of myeloid cell differentiation during cancer is crucial to counteract their pro-tumourigenic role. Bone marrow (BM) haematopoiesis is a tightly regulated process for the production of all immune cells in accordance to tissue needs14. Myeloid cells differentiate during haematopoiesis from multipotent haematopoietic stem and progenitor cells (HSPCs)15-17. HSPCs can sense inflammatory signals from the periphery during infections18-21 or inflammatory disorders22-27. In these settings, HSPC expansion is associated with increased myeloid differentiation28,29. During carcinogenesis, the elevation of haematopoietic growth factors supports the expansion and differentiation of committed myeloid progenitors5,30. However, it is unclear whether cancer-related inflammation also triggers demand-adapted haematopoiesis at the level of multipotent HSPCs. In the BM, HSPCs reside within the haematopoietic niche which delivers HSC maintenance and differentiation cues31-35. Mesenchymal stem cells (MSCs) are a major cellular component of the BM niche and contribute to HSC homeostasis36-41. Modifications of MSCs in systemic disorders have been associated with HSC differentiation towards myeloid cells22,42. It is unknown if MSCs are regulated in the context of solid tumours and if their myeloid supportive activity is impacted by cancer-induced systemic changes. Here, using unbiased transcriptomic analysis and in situ imaging of HSCs and the BM niche during breast cancer, we show that both HSCs and MSCs are transcriptionally and spatially modified. We demonstrate that breast tumour can distantly remodel the cellular cross-talks in the BM niche leading to increased myelopoiesis.


Subject(s)
Bone Marrow , Breast Neoplasms , Humans , Female , Breast Neoplasms/pathology , Hematopoietic Stem Cells/metabolism , Multipotent Stem Cells/metabolism , Cell Differentiation , Stem Cell Niche , Bone Marrow Cells
SELECTION OF CITATIONS
SEARCH DETAIL