Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 114
1.
Sci Immunol ; 9(94): eadi1023, 2024 Apr 12.
Article En | MEDLINE | ID: mdl-38608038

The development of dendritic cells (DCs), including antigen-presenting conventional DCs (cDCs) and cytokine-producing plasmacytoid DCs (pDCs), is controlled by the growth factor Flt3 ligand (Flt3L) and its receptor Flt3. We genetically dissected Flt3L-driven DC differentiation using CRISPR-Cas9-based screening. Genome-wide screening identified multiple regulators of DC differentiation including subunits of TSC and GATOR1 complexes, which restricted progenitor growth but enabled DC differentiation by inhibiting mTOR signaling. An orthogonal screen identified the transcriptional repressor Trim33 (TIF-1γ) as a regulator of DC differentiation. Conditional targeting in vivo revealed an essential role of Trim33 in the development of all DCs, but not of monocytes or granulocytes. In particular, deletion of Trim33 caused rapid loss of DC progenitors, pDCs, and the cross-presenting cDC1 subset. Trim33-deficient Flt3+ progenitors up-regulated pro-inflammatory and macrophage-specific genes but failed to induce the DC differentiation program. Collectively, these data elucidate mechanisms that control Flt3L-driven differentiation of the entire DC lineage and identify Trim33 as its essential regulator.


Chorea , Cell Differentiation , Cytokines , Dendritic Cells
2.
Proc Natl Acad Sci U S A ; 121(4): e2317929121, 2024 Jan 23.
Article En | MEDLINE | ID: mdl-38227649

The hierarchical model of hematopoiesis posits that self-renewing, multipotent hematopoietic stem cells (HSCs) give rise to all blood cell lineages. While this model accounts for hematopoiesis in transplant settings, its applicability to steady-state hematopoiesis remains to be clarified. Here, we used inducible clonal DNA barcoding of endogenous adult HSCs to trace their contribution to major hematopoietic cell lineages in unmanipulated animals. While the majority of barcodes were unique to a single lineage, we also observed frequent barcode sharing between multiple lineages, specifically between lymphocytes and myeloid cells. These results suggest that both single-lineage and multilineage contributions by HSCs collectively drive continuous hematopoiesis, and highlight a close relationship of myeloid and lymphoid development.


Adult Stem Cells , Hematopoietic Stem Cells , Animals , Cell Differentiation , Hematopoiesis/genetics , Cell Lineage/genetics
3.
J Immunol ; 211(10): 1475-1480, 2023 11 15.
Article En | MEDLINE | ID: mdl-37800687

Autoantibodies to chromatin and dsDNA are a hallmark of systemic lupus erythematosus (SLE). In a mouse model of monogenic human SLE caused by DNASE1L3 deficiency, the anti-DNA response is dependent on endosomal nucleic acid-sensing TLRs TLR7 and TLR9. In this study, we report that this response also required TLR2, a surface receptor for microbial products that is primarily expressed on myeloid cells. Cell transfers into lymphopenic DNASE1L3-deficient mice showed that TLR2 was required for anti-DNA Ab production by lymphocytes. TLR2 was detectably expressed on B cells and facilitated the production of IL-6 by B cells activated in the presence of microbial products. Accordingly, treatment with broad-spectrum antibiotics or Ab-mediated blockade of IL-6 delayed the anti-DNA response in DNASE1L3-deficient mice. These studies reveal an unexpected B cell-intrinsic role of TLR2 in systemic autoreactivity to DNA, and they suggest that microbial products may synergize with self-DNA in the activation of autoreactive B cells in SLE.


Lupus Erythematosus, Systemic , Toll-Like Receptor 2 , Mice , Animals , Humans , Interleukin-6 , B-Lymphocytes , Autoantibodies , Antibodies, Antinuclear , DNA
4.
Nat Immunol ; 24(8): 1265-1280, 2023 08.
Article En | MEDLINE | ID: mdl-37414907

High-dimensional approaches have revealed heterogeneity amongst dendritic cells (DCs), including a population of transitional DCs (tDCs) in mice and humans. However, the origin and relationship of tDCs to other DC subsets has been unclear. Here we show that tDCs are distinct from other well-characterized DCs and conventional DC precursors (pre-cDCs). We demonstrate that tDCs originate from bone marrow progenitors shared with plasmacytoid DCs (pDCs). In the periphery, tDCs contribute to the pool of ESAM+ type 2 DCs (DC2s), and these DC2s have pDC-related developmental features. Different from pre-cDCs, tDCs have less turnover, capture antigen, respond to stimuli and activate antigen-specific naïve T cells, all characteristics of differentiated DCs. Different from pDCs, viral sensing by tDCs results in IL-1ß secretion and fatal immune pathology in a murine coronavirus model. Our findings suggest that tDCs are a distinct pDC-related subset with a DC2 differentiation potential and unique proinflammatory function during viral infections.


Bone Marrow , Dendritic Cells , Animals , Mice , Antiviral Agents , Bone Marrow/immunology , Cell Differentiation , Dendritic Cells/classification , Dendritic Cells/immunology
5.
Blood ; 142(5): 460-476, 2023 08 03.
Article En | MEDLINE | ID: mdl-37267505

The chromosome 9p21 locus comprises several tumor suppressor genes including MTAP, CDKN2A, and CDKN2B, and its homo- or heterozygous deletion is associated with reduced survival in multiple cancer types. We report that mice with germ line monoallelic deletion or induced biallelic deletion of the 9p21-syntenic locus (9p21s) developed a fatal myelodysplastic syndrome/myeloproliferative neoplasm (MDS/MPN)-like disease associated with aberrant trabecular bone formation and/or fibrosis in the bone marrow (BM). Reciprocal BM transfers and conditional targeting of 9p21s suggested that the disease originates in the BM stroma. Single-cell analysis of 9p21s-deficient BM stroma revealed the expansion of chondrocyte and osteogenic precursors, reflected in increased osteogenic differentiation in vitro. It also showed reduced expression of factors maintaining hematopoietic stem/progenitor cells, including Cxcl12. Accordingly, 9p21s-deficient mice showed reduced levels of circulating Cxcl12 and concomitant upregulation of the profibrotic chemokine Cxcl13 and the osteogenesis- and fibrosis-related multifunctional glycoprotein osteopontin/Spp1. Our study highlights the potential of mutations in the BM microenvironment to drive MDS/MPN-like disease.


Bone Marrow , Osteogenesis , Mice , Animals , Bone Marrow/pathology , Hematopoietic Stem Cells/metabolism , Genes, Tumor Suppressor , Cell Differentiation
6.
Proc Natl Acad Sci U S A ; 120(22): e2302019120, 2023 05 30.
Article En | MEDLINE | ID: mdl-37216517

Mammalian aging is associated with multiple defects of hematopoiesis, most prominently with the impaired development of T and B lymphocytes. This defect is thought to originate in hematopoietic stem cells (HSCs) of the bone marrow, specifically due to the age-dependent accumulation of HSCs with preferential megakaryocytic and/or myeloid potential ("myeloid bias"). Here, we tested this notion using inducible genetic labeling and tracing of HSCs in unmanipulated animals. We found that the endogenous HSC population in old mice shows reduced differentiation into all lineages including lymphoid, myeloid, and megakaryocytic. Single-cell RNA sequencing and immunophenotyping (CITE-Seq) showed that HSC progeny in old animals comprised balanced lineage spectrum including lymphoid progenitors. Lineage tracing using the aging-induced HSC marker Aldh1a1 confirmed the low contribution of old HSCs across all lineages. Competitive transplantations of total bone marrow cells with genetically marked HSCs revealed that the contribution of old HSCs was reduced, but compensated by other donor cells in myeloid cells but not in lymphocytes. Thus, the HSC population in old animals becomes globally decoupled from hematopoiesis, which cannot be compensated in lymphoid lineages. We propose that this partially compensated decoupling, rather than myeloid bias, is the primary cause of the selective impairment of lymphopoiesis in older mice.


Aging , Hematopoietic Stem Cells , Mice , Animals , Cell Lineage , Cell Differentiation , Bone Marrow , Hematopoiesis , Mammals
7.
J Exp Med ; 220(6)2023 06 05.
Article En | MEDLINE | ID: mdl-36928522

Extracellular DNase DNASE1L3 maintains tolerance to self-DNA in humans and mice, whereas the role of its homolog DNASE1 remains controversial, and the overall function of secreted DNases in immunity is unclear. We report that deletion of murine DNASE1 neither caused autoreactivity in isolation nor exacerbated lupus-like disease in DNASE1L3-deficient mice. However, combined deficiency of DNASE1 and DNASE1L3 rendered mice susceptible to bloodstream infection with Staphylococcus aureus. DNASE1/DNASE1L3 double-deficient mice mounted a normal innate response to S. aureus and did not accumulate neutrophil extracellular traps (NETs). However, their kidneys manifested severe pathology, increased bacterial burden, and biofilm-like bacterial lesions that contained bacterial DNA and excluded neutrophils. Furthermore, systemic administration of recombinant DNASE1 protein during S. aureus infection rescued the mortality of DNase-deficient mice and ameliorated the disease in wild-type mice. Thus, DNASE1 and DNASE1L3 jointly facilitate the control of bacterial infection by digesting extracellular microbial DNA in biofilms, suggesting the original evolutionary function of secreted DNases as antimicrobial agents.


Biofilms , Endodeoxyribonucleases , Sepsis , Staphylococcal Infections , Animals , Mice , Deoxyribonuclease I/genetics , Deoxyribonuclease I/metabolism , Deoxyribonucleases/metabolism , DNA/metabolism , Endodeoxyribonucleases/metabolism , Extracellular Traps/metabolism , Mammals/genetics , Mammals/metabolism , Staphylococcal Infections/prevention & control , Staphylococcus aureus/metabolism
8.
Methods Mol Biol ; 2618: 109-119, 2023.
Article En | MEDLINE | ID: mdl-36905512

Dendritic cells (DCs) comprise a heterogeneous population of antigen (Ag)-presenting cells that play a critical role in both innate and adaptive immunity. DCs orchestrate protective responses against pathogens and tumors while mediating tolerance to host tissues. Evolutionary conservation between species has allowed the successful use of murine models to identify and characterize DC types and functions relevant to human health. Among DCs, type 1 classical DCs (cDC1) are uniquely capable of inducing antitumor responses and therefore present a promising therapeutic target. However, the rarity of DCs, particularly cDC1, limits the number of cells that can be isolated for study. Despite significant effort, progress in the field has been hampered by inadequate methods to produce large quantities of functionally mature DCs in vitro. To overcome this challenge, we developed a culture system in which mouse primary bone marrow cells are cocultured with OP9 stromal cells expressing Notch ligand Delta-like 1 (OP9-DL1) to produce CD8α+ DEC205+ XCR1+ cDC1 (Notch cDC1). This novel method provides a valuable tool to facilitate the generation of unlimited cDC1 for functional studies and translational applications such as antitumor vaccination and immunotherapy.


Bone Marrow , Dendritic Cells , Mice , Humans , Animals , Bone Marrow Cells , Immunity , Coculture Techniques
10.
Nat Immunol ; 24(1): 84-95, 2023 01.
Article En | MEDLINE | ID: mdl-36543959

In inflamed tissues, monocytes differentiate into macrophages (mo-Macs) or dendritic cells (mo-DCs). In chronic nonresolving inflammation, mo-DCs are major drivers of pathogenic events. Manipulating monocyte differentiation would therefore be an attractive therapeutic strategy. However, how the balance of mo-DC versus mo-Mac fate commitment is regulated is not clear. In the present study, we show that the transcriptional repressors ETV3 and ETV6 control human monocyte differentiation into mo-DCs. ETV3 and ETV6 inhibit interferon (IFN)-stimulated genes; however, their action on monocyte differentiation is independent of IFN signaling. Instead, we find that ETV3 and ETV6 directly repress mo-Mac development by controlling MAFB expression. Mice deficient for Etv6 in monocytes have spontaneous expression of IFN-stimulated genes, confirming that Etv6 regulates IFN responses in vivo. Furthermore, these mice have impaired mo-DC differentiation during inflammation and reduced pathology in an experimental autoimmune encephalomyelitis model. These findings provide information about the molecular control of monocyte fate decision and identify ETV6 as a therapeutic target to redirect monocyte differentiation in inflammatory disorders.


Dendritic Cells , Monocytes , Animals , Humans , Mice , Cell Differentiation , Cells, Cultured , Inflammation/metabolism , Macrophages , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , ETS Translocation Variant 6 Protein
11.
J Immunol ; 209(10): 1906-1917, 2022 11 15.
Article En | MEDLINE | ID: mdl-36426957

Rheumatoid arthritis (RA) is characterized by autoimmune joint destruction with debilitating consequences. Despite treatment advancements with biologic therapies, a significant proportion of RA patients show an inadequate clinical response, and restoration of immune self-tolerance represents an unmet therapeutic need. We have previously described a tolerogenic phenotype of plasmacytoid dendritic cells (pDCs) in RA patients responding to anti-TNF-α agents. However, the molecular mechanisms involved in tolerogenic reprogramming of pDCs in RA remain elusive. In this study, guided by transcriptomic analysis of CD303+CD123+ pDCs from RA patients in remission, we revealed enhanced expression of IL-6R and its downstream signaling compared with healthy pDCs. Functional assessment demonstrated that IL-6R engagement resulted in marked reduction of TNF-α secretion by pDCs whereas intracellular TNF-α was significantly increased. Accordingly, pharmacologic inhibition of IL-6R signaling restored TNF-α secretion levels by pDCs. Mechanistic analysis demonstrated impaired activity and decreased lysosomal degradation of ADAM17 (a disintegrin and metalloproteinase 17) sheddase in pDCs, which is essential for TNF-α cleavage. Importantly, reduction of TNF-α secretion by IL-6-treated pDCs attenuated the inflammatory potential of RA patient-derived synovial fibroblasts. Collectively, these findings position pDCs as an important source of TNF-α in RA pathogenesis and unravel an anti-inflammatory mechanism of IL-6 by limiting the pDC-derived TNF-α secretion.


Arthritis, Rheumatoid , Interleukin-6 , Humans , Tumor Necrosis Factor Inhibitors , Dendritic Cells , Signal Transduction , Tumor Necrosis Factor-alpha
12.
Immunity ; 55(3): 405-422.e11, 2022 03 08.
Article En | MEDLINE | ID: mdl-35180378

Developmental origins of dendritic cells (DCs) including conventional DCs (cDCs, comprising cDC1 and cDC2 subsets) and plasmacytoid DCs (pDCs) remain unclear. We studied DC development in unmanipulated adult mice using inducible lineage tracing combined with clonal DNA "barcoding" and single-cell transcriptome and phenotype analysis (CITE-seq). Inducible tracing of Cx3cr1+ hematopoietic progenitors in the bone marrow showed that they simultaneously produce all DC subsets including pDCs, cDC1s, and cDC2s. Clonal tracing of hematopoietic stem cells (HSCs) and of Cx3cr1+ progenitors revealed clone sharing between cDC1s and pDCs, but not between the two cDC subsets or between pDCs and B cells. Accordingly, CITE-seq analyses of differentiating HSCs and Cx3cr1+ progenitors identified progressive stages of pDC development including Cx3cr1+ Ly-6D+ pro-pDCs that were distinct from lymphoid progenitors. These results reveal the shared origin of pDCs and cDCs and suggest a revised scheme of DC development whereby pDCs share clonal relationship with cDC1s.


B-Lymphocytes , Dendritic Cells , Animals , Cell Count , Chorea , Hematopoietic Stem Cells , Mice
13.
Front Immunol ; 12: 757231, 2021.
Article En | MEDLINE | ID: mdl-34630435

Dendritic cells (DCs) are composed of multiple lineages of hematopoietic cells and orchestrate immune responses upon detecting the danger and inflammatory signals associated with pathogen and damaged tissues. Under steady-state, DCs are maintained at limited numbers and the functionally quiescent status. While it is known that a fine balance in the DC homeostasis and activation status is also important to prevent autoimmune diseases and hyperinflammation, mechanisms that control DC development and activation under stead-state remain not fully understood. Here we show that DC-specific ablation of CBL and CBL-B (CBL-/-CBL-B-/-) leads to spontaneous liver inflammation and fibrosis and early death of the mice. The mutant mice have a marked expansion of classic CD8α+/CD103+ DCs (cDC1s) in peripheral lymphoid organs and the liver. These DCs exhibit atypical activation phenotypes characterized by an increased production of inflammatory cytokines and chemokines but not the cell surface MHC-II and costimulatory ligands. While the mutant mice also have massive T cell activation, lymphocytes are not required for the disease development. The CBL-/-CBL-B-/- mutation enhances FLT3-mTOR signaling, due to defective FLT3 ubiquitination and degradation. Blockade of FLT3-mTOR signaling normalizes the homeostasis of cDC1s and attenuates liver inflammation. Our result thus reveals a critical role of CBLs in the maintenance of DC homeostasis and immune quiescence. This regulation could be relevant to liver inflammatory diseases and fibrosis in humans.


Adaptor Proteins, Signal Transducing/physiology , Dendritic Cells/immunology , Proto-Oncogene Proteins c-cbl/physiology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Antigen Presentation , Cell Division , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Hepatitis, Autoimmune/genetics , Hepatitis, Autoimmune/immunology , Homeostasis , Lymphocyte Subsets/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Point Mutation , Proto-Oncogene Proteins c-akt/physiology , Proto-Oncogene Proteins c-cbl/deficiency , Proto-Oncogene Proteins c-cbl/genetics , Sirolimus/pharmacology , fms-Like Tyrosine Kinase 3/physiology
14.
Nature ; 595(7868): 578-584, 2021 07.
Article En | MEDLINE | ID: mdl-34135508

Macrophages have a key role in shaping the tumour microenvironment (TME), tumour immunity and response to immunotherapy, which makes them an important target for cancer treatment1,2. However, modulating macrophages has proved extremely difficult, as we still lack a complete understanding of the molecular and functional diversity of the tumour macrophage compartment. Macrophages arise from two distinct lineages. Tissue-resident macrophages self-renew locally, independent of adult haematopoiesis3-5, whereas short-lived monocyte-derived macrophages arise from adult haematopoietic stem cells, and accumulate mostly in inflamed lesions1. How these macrophage lineages contribute to the TME and cancer progression remains unclear. To explore the diversity of the macrophage compartment in human non-small cell lung carcinoma (NSCLC) lesions, here we performed single-cell RNA sequencing of tumour-associated leukocytes. We identified distinct populations of macrophages that were enriched in human and mouse lung tumours. Using lineage tracing, we discovered that these macrophage populations differ in origin and have a distinct temporal and spatial distribution in the TME. Tissue-resident macrophages accumulate close to tumour cells early during tumour formation to promote epithelial-mesenchymal transition and invasiveness in tumour cells, and they also induce a potent regulatory T cell response that protects tumour cells from adaptive immunity. Depletion of tissue-resident macrophages reduced the numbers and altered the phenotype of regulatory T cells, promoted the accumulation of CD8+ T cells and reduced tumour invasiveness and growth. During tumour growth, tissue-resident macrophages became redistributed at the periphery of the TME, which becomes dominated by monocyte-derived macrophages in both mouse and human NSCLC. This study identifies the contribution of tissue-resident macrophages to early lung cancer and establishes them as a target for the prevention and treatment of early lung cancer lesions.


Carcinogenesis , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Macrophages/immunology , Tumor Microenvironment , Animals , CD8-Positive T-Lymphocytes/immunology , Epithelial-Mesenchymal Transition , Female , Humans , Male , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness , T-Lymphocytes, Regulatory/immunology
15.
Sci Adv ; 7(21)2021 05.
Article En | MEDLINE | ID: mdl-34020954

Inflammation is known to adversely affect adult neurogenesis, wherein the source of inflammation is largely thought to be extraneous to the neurogenic niche. Here, we demonstrate that the adult hippocampal neural progenitors harbor an inflammatory potential that is proactively suppressed by transcription factor 4 (Tcf4). Deletion of Tcf4 in hippocampal nestin-expressing progenitors causes loss of proliferative capacity and acquisition of myeloid inflammatory properties. This transformation abolishes their differentiation potential and causes production of detrimental factors that adversely affect niche cells, causing inflammation in the dentate gyrus. Thus, on one hand, Tcf4 deletion causes abrogation of proliferative progenitors leading to reduction of adult neurogenesis, while on the other, their accompanying inflammatory transformation inflicts inflammation in the niche. Taken together, we provide the first evidence for a latent inflammatory potential of adult hippocampal neural progenitors and identify Tcf4 as a critical regulator that facilitates adult neurogenesis via proactive suppression of this detrimental potential.


Dentate Gyrus , Neural Stem Cells , Transcription Factor 4 , Animals , Inflammation/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis , Transcription Factor 4/genetics
16.
Sci Immunol ; 6(58)2021 04 02.
Article En | MEDLINE | ID: mdl-33811059

Plasmacytoid dendritic cells (pDCs) can rapidly produce interferons and other soluble factors in response to extracellular viruses or virus mimics such as CpG-containing DNA. pDCs can also recognize live cells infected with certain RNA viruses, but the relevance and functional consequences of such recognition remain unclear. We studied the response of primary DCs to the prototypical persistent DNA virus, human cytomegalovirus (CMV). Human pDCs produced high amounts of type I interferon (IFN-I) when incubated with live CMV-infected fibroblasts but not with free CMV; the response involved integrin-mediated adhesion, transfer of DNA-containing virions to pDCs, and the recognition of DNA through TLR9. Compared with transient polyfunctional responses to CpG or free influenza virus, pDC response to CMV-infected cells was long-lasting, dominated by the production of IFN-I and IFN-III, and lacked diversification into functionally distinct populations. Similarly, pDC activation by influenza-infected lung epithelial cells was highly efficient, prolonged, and dominated by interferon production. Prolonged pDC activation by CMV-infected cells facilitated the activation of natural killer cells critical for CMV control. Last, patients with CMV viremia harbored phenotypically activated pDCs and increased circulating IFN-I and IFN-III. Thus, recognition of live infected cells is a mechanism of virus detection by pDCs that elicits a unique antiviral immune response.


Dendritic Cells/immunology , Fibroblasts/immunology , Interferon Type I/metabolism , Interferons/metabolism , Cells, Cultured , Coculture Techniques , CpG Islands/immunology , Cytomegalovirus/immunology , Dendritic Cells/metabolism , Fibroblasts/virology , Host Microbial Interactions/immunology , Humans , Primary Cell Culture , Interferon Lambda
17.
J Exp Med ; 218(5)2021 05 03.
Article En | MEDLINE | ID: mdl-33783474

Antibodies to double-stranded DNA (dsDNA) are prevalent in systemic lupus erythematosus (SLE), particularly in patients with lupus nephritis, yet the nature and regulation of antigenic cell-free DNA (cfDNA) are poorly understood. Null mutations in the secreted DNase DNASE1L3 cause human monogenic SLE with anti-dsDNA autoreactivity. We report that >50% of sporadic SLE patients with nephritis manifested reduced DNASE1L3 activity in circulation, which was associated with neutralizing autoantibodies to DNASE1L3. These patients had normal total plasma cfDNA levels but showed accumulation of cfDNA in circulating microparticles. Microparticle-associated cfDNA contained a higher fraction of longer polynucleosomal cfDNA fragments, which bound autoantibodies with higher affinity than mononucleosomal fragments. Autoantibodies to DNASE1L3-sensitive antigens on microparticles were prevalent in SLE nephritis patients and correlated with the accumulation of cfDNA in microparticles and with disease severity. DNASE1L3-sensitive antigens included DNA-associated proteins such as HMGB1. Our results reveal autoantibody-mediated impairment of DNASE1L3 activity as a common nongenetic mechanism facilitating anti-dsDNA autoreactivity in patients with severe sporadic SLE.


Antibodies, Antinuclear/immunology , Autoantibodies/immunology , DNA/immunology , Endodeoxyribonucleases/immunology , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/immunology , Adult , Animals , Antibodies, Antinuclear/blood , Autoantibodies/blood , Cell-Derived Microparticles/immunology , Cell-Derived Microparticles/metabolism , Cell-Free Nucleic Acids/blood , Cell-Free Nucleic Acids/immunology , Child , Endodeoxyribonucleases/blood , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Female , HEK293 Cells , HMGB1 Protein/immunology , HMGB1 Protein/metabolism , Humans , Lupus Erythematosus, Systemic/metabolism , Lupus Nephritis/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Severity of Illness Index
18.
Proc Natl Acad Sci U S A ; 117(49): 31331-31342, 2020 12 08.
Article En | MEDLINE | ID: mdl-33214146

Antigen-presenting conventional dendritic cells (cDCs) are broadly divided into type 1 and type 2 subsets that further adapt their phenotype and function to perform specialized tasks in the immune system. The precise signals controlling tissue-specific adaptation and differentiation of cDCs are currently poorly understood. We found that mice deficient in the Ste20 kinase Thousand and One Kinase 3 (TAOK3) lacked terminally differentiated ESAM+ CD4+ cDC2s in the spleen and failed to prime CD4+ T cells in response to allogeneic red-blood-cell transfusion. These NOTCH2- and ADAM10-dependent cDC2s were absent selectively in the spleen, but not in the intestine of Taok3-/- and CD11c-cre Taok3fl/fl mice. The loss of splenic ESAM+ cDC2s was cell-intrinsic and could be rescued by conditional overexpression of the constitutively active NOTCH intracellular domain in CD11c-expressing cells. Therefore, TAOK3 controls the terminal differentiation of NOTCH2-dependent splenic cDC2s.


Cell Differentiation , Dendritic Cells/cytology , Dendritic Cells/enzymology , Protein Kinases/metabolism , Receptor, Notch2/metabolism , Spleen/cytology , Animals , Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/immunology , Gene Expression Regulation , Intestine, Small/metabolism , Mice, Inbred C57BL , Phenotype , Protein Domains , Protein Kinases/deficiency , Receptor, Notch2/chemistry , Signal Transduction
19.
Am J Hum Genet ; 107(5): 882-894, 2020 11 05.
Article En | MEDLINE | ID: mdl-33022220

Plasma DNA fragmentomics is an emerging area in cell-free DNA diagnostics and research. In murine models, it has been shown that the extracellular DNase, DNASE1L3, plays a role in the fragmentation of plasma DNA. In humans, DNASE1L3 deficiency causes familial monogenic systemic lupus erythematosus with childhood onset and anti-dsDNA reactivity. In this study, we found that human patients with DNASE1L3 disease-associated gene variations showed aberrations in size and a reduction of a "CC" end motif of plasma DNA. Furthermore, we demonstrated that DNA from DNASE1L3-digested cell nuclei showed a median length of 153 bp with CC motif frequencies resembling plasma DNA from healthy individuals. Adeno-associated virus-based transduction of Dnase1l3 into Dnase1l3-deficient mice restored the end motif profiles to those seen in the plasma DNA of wild-type mice. Our findings demonstrate that DNASE1L3 is an important player in the fragmentation of plasma DNA, which appears to act in a cell-extrinsic manner to regulate plasma DNA size and motif frequency.


DNA/genetics , Endodeoxyribonucleases/genetics , Lupus Erythematosus, Systemic/genetics , Mutation , Animals , Case-Control Studies , DNA/blood , DNA Fragmentation , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Endodeoxyribonucleases/deficiency , Endodeoxyribonucleases/metabolism , Genetic Therapy , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Lupus Erythematosus, Systemic/enzymology , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Transgenic , Substrate Specificity , Transduction, Genetic
20.
Cell Stem Cell ; 27(2): 336-345.e4, 2020 08 06.
Article En | MEDLINE | ID: mdl-32589864

Adult mammalian hematopoietic stem cells (HSCs) reside in the bone marrow (BM) but can be mobilized into blood for use in transplantation. HSCs interact with BM niche cells that produce growth factor c-Kit ligand (Kitl/SCF) and chemokine CXCL12, and were thought to be static and sessile. We used two-photon laser scanning microscopy to visualize genetically labeled HSCs in the BM of live mice for several hours. The majority of HSCs showed a dynamic non-spherical morphology and significant motility, undergoing slow processive motion interrupted by short stretches of confined motion. HSCs moved in the perivascular space and showed intermittent close contacts with SCF-expressing perivascular stromal cells. In contrast, mobilization-inducing blockade of CXCL12 receptor CXCR4 and integrins rapidly abrogated HSC motility and shape dynamics in real time. Our results reveal an unexpectedly dynamic nature of HSC residence in the BM and interaction with the SCF+ stromal niche, which is disrupted during HSC mobilization.


Bone Marrow , Hematopoietic Stem Cells , Animals , Bone Marrow Cells , Cell Movement , Chemokine CXCL12 , Intravital Microscopy , Mice , Stem Cell Niche
...