Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 423
Filter
1.
Sci Adv ; 10(23): eadn8963, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38838144

ABSTRACT

Nucleoporins, the components of nuclear pore complexes (NPCs), can play cell type- and tissue-specific functions. Yet, the physiological roles and mechanisms of action for most NPC components have not yet been established. We report that Nup358, a nucleoporin linked to several myeloid disorders, is required for the developmental progression of early myeloid progenitors. We found that Nup358 ablation in mice results in the loss of myeloid-committed progenitors and mature myeloid cells and the accumulation of myeloid-primed multipotent progenitors (MPPs) in bone marrow. Accumulated MPPs in Nup358 knockout mice are greatly restricted to megakaryocyte/erythrocyte-biased MPP2, which fail to progress into committed myeloid progenitors. Mechanistically, we found that Nup358 is required for histone deacetylase 3 (HDAC3) nuclear import and function in MPP2 cells and established that this nucleoporin regulates HDAC3 nuclear translocation in a SUMOylation-independent manner. Our study identifies a critical function for Nup358 in myeloid-primed MPP2 differentiation and uncovers an unexpected role for NPCs in the early steps of myelopoiesis.


Subject(s)
Cell Differentiation , Histone Deacetylases , Mice, Knockout , Nuclear Pore Complex Proteins , Animals , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Nuclear Pore Complex Proteins/metabolism , Nuclear Pore Complex Proteins/genetics , Mice , Molecular Chaperones/metabolism , Molecular Chaperones/genetics , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/cytology , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Multipotent Stem Cells/metabolism , Multipotent Stem Cells/cytology , Myeloid Cells/metabolism , Myeloid Cells/cytology , Sumoylation , Myelopoiesis/genetics
2.
J Leukoc Biol ; 111(1): 113-122, 2022 01.
Article in English | MEDLINE | ID: mdl-33857341

ABSTRACT

Bone marrow is a hematopoietic site harboring multiple populations of myeloid cells in different stages of differentiation. Murine bone marrow eosinophils are traditionally identified by Siglec-F(+) staining using flow cytometry, whereas neutrophils are characterized by Ly6G(+) expression. However, using flow cytometry to characterize bone marrow hematopoietic cells in wild-type mice, we found substantial gray areas in identification of these cells. Siglec-F(+) mature eosinophil population constituted only a minority of bone marrow Lin(+)CD45(+) pool (5%). A substantial population of Siglec-F(-) cells was double positive for neutrophil marker Ly6G and eosinophil lineage marker, IL-5Rα. This granulocyte population with mixed neutrophil and eosinophil characteristics is typically attributable to neutrophil pool based on neutral granule staining and expression of Ly6G and myeloid peroxidase. It is distinct from Lineage(-) myeloid progenitors or Siglec-F(+)Ly6G(+) maturing eosinophil precursors, and can be accurately identified by Lineage(+) staining and positive expression of markers IL-5Rα and Ly6G. At 15-50% of all CD45(+) hematopoietic cells in adult mice (percentage varies by sex and age), this is a surprisingly dominant population, which increases with age in both male and female mice. RNA-seq characterization of these cells revealed a complex immune profile and the capacity to secrete constituents of the extracellular matrix. When sorted from bone marrow, these resident cells had neutrophilic phenotype but readily acquired all characteristics of eosinophils when cultured with G-CSF or IL-5, including expression of Siglec-F and granular proteins (Epx, Mbp). Surprisingly, these cells were also able to differentiate into Ly6C(+) monocytes when cultured with M-CSF. Herein described is the discovery of an unexpected hematopoietic flexibility of a dominant population of multipotent myeloid cells, typically categorized as neutrophils, but with the previously unknown plasticity to contribute to mature pools of eosinophils and monocytes.


Subject(s)
Antigens, Ly/analysis , Eosinophils/cytology , Interleukin-5 Receptor alpha Subunit/analysis , Monocytes/cytology , Myeloid Progenitor Cells/cytology , Neutrophils/cytology , Animals , Bone Marrow Cells/cytology , Cell Differentiation , Cells, Cultured , Female , Leukopoiesis , Male , Mice, Inbred BALB C
3.
Stem Cell Reports ; 16(12): 2871-2886, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34798065

ABSTRACT

Hematopoietic cells differentiate through several progenitors in a hierarchical manner, and recent single-cell analyses have revealed substantial heterogeneity within each progenitor. Although common myeloid progenitors (CMPs) are defined as a multipotent cell population that can differentiate into granulocyte-monocyte progenitors (GMPs) and megakaryocyte-erythrocyte progenitors (MEPs), and GMPs generate neutrophils and monocytes, these myeloid progenitors must contain some lineage-committed progenitors. Through gene expression analysis at single-cell levels, we identified CD62L as a marker to reveal the heterogeneity. We confirmed that CD62L-negative CMPs represent "bona fide" CMPs, whereas CD62L-high CMPs are mostly restricted to GMP potentials both in mice and humans. In addition, we identified CD62L-negative GMPs as the most immature subsets in GMPs and Ly6C+CD62L-intermediate and Ly6C+CD62L-high GMPs are skewed to neutrophil and monocyte differentiation in mice, respectively. Our findings contribute to more profound understanding about the mechanism of myeloid differentiation.


Subject(s)
Cell Lineage , L-Selectin/metabolism , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Animals , Cell Differentiation , Gene Expression Profiling , Gene Expression Regulation , Humans , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice, Inbred C57BL , Monocytes/cytology , Monocytes/metabolism , Neutrophils/cytology , Neutrophils/metabolism
4.
Mutat Res ; 823: 111756, 2021.
Article in English | MEDLINE | ID: mdl-34153743

ABSTRACT

We investigated the effects of 50 Hz extremely low-frequency magnetic fields (MFs) on gene expression related to the circadian rhythm or DNA damage signaling and whether these fields modify DNA damage repair rate after bleomycin treatment. Murine FDC-P1 hematopoietic cells were exposed for different durations (15 min, 2 h, 12 h, and 24 h) to either 200 µT MFs or sham-exposures. Cells were then collected for comet assay or real-time PCR to determine immediate DNA damage level and circadian rhythm gene expression, respectively. To assess DNA-damage signaling and DNA repair rate, the cells were subsequently treated with 20 µg/mL bleomycin for 1 h and then either assayed immediately or allowed to repair their DNA for 1 or 2 h. We found that circadian rhythm-related genes were upregulated after 12 h of MF exposure and downregulated after 24 h of MF exposure, but none of the affected genes were core genes controlling the circadian rhythm. In addition, we found that the repair rate for bleomycin-induced damage was only decreased after MF exposure for 24 h. In conclusion, our findings suggest that the effects of MFs are duration-dependent; they were observed predominantly after long exposures.


Subject(s)
Circadian Clocks/drug effects , Circadian Rhythm Signaling Peptides and Proteins/genetics , DNA Repair , Magnetic Fields/adverse effects , Monocytes/drug effects , Mutation , Animals , Bleomycin/pharmacology , Cell Differentiation , Cell Line , Circadian Clocks/genetics , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Comet Assay , DNA Damage , Gene Expression/drug effects , Mice , Monocytes/cytology , Monocytes/metabolism , Mutagens/pharmacology , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/drug effects , Myeloid Progenitor Cells/metabolism , Time Factors
5.
Eur J Immunol ; 51(9): 2237-2250, 2021 09.
Article in English | MEDLINE | ID: mdl-34107067

ABSTRACT

Early embryonic hematopoiesis in mammals is defined by three successive waves of hematopoietic progenitors which exhibit a distinct hematopoietic potential and provide continuous support for the development of the embryo and adult organism. Although the functional importance of each of these waves has been analyzed, their spatio-temporal overlap and the lack of wave-specific markers hinders the accurate separation and assessment of their functional roles during early embryogenesis. We have recently shown that TLR2, in combination with c-kit, represents the earliest signature of emerging precursors of the second hematopoietic wave, erythro-myeloid precursors (EMPs). Since the onset of Tlr2 expression distinguishes EMPs from primitive progenitors which coexist in the yolk sac from E7.5, we generated a novel transgenic "knock in" mouse model, Tlr2Dtr , suitable for inducible targeted depletion of TLR2+ EMPs. In this model, the red fluorescent protein and diphtheria toxin receptor sequences are linked via a P2A sequence and inserted into the Tlr2 locus before its stop codon. We show that a timely controlled deletion of TLR2+ EMPs in Tlr2Dtr embryos results in a marked decrease in both erythroid as well as myeloid lineages and, consequently, in embryonic lethality peaking before E13.5. These findings validate the importance of EMPs in embryonic development.


Subject(s)
Embryo, Mammalian/pathology , Embryonic Development/genetics , Hematopoiesis/genetics , Myeloid Progenitor Cells/cytology , Toll-Like Receptor 2/genetics , Animals , Embryo, Mammalian/embryology , Erythrocytes/cytology , Hematopoiesis/physiology , Macrophages/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic
6.
Sci Rep ; 11(1): 10736, 2021 05 24.
Article in English | MEDLINE | ID: mdl-34031489

ABSTRACT

The transmembrane aminopeptidase CD13 is highly expressed in cells of the myeloid lineage, regulates dynamin-dependent receptor endocytosis and recycling and is a necessary component of actin cytoskeletal organization. Here, we show that CD13-deficient mice present a low bone density phenotype with increased numbers of osteoclasts per bone surface, but display a normal distribution of osteoclast progenitor populations in the bone marrow and periphery. In addition, the bone formation and mineral apposition rates are similar between genotypes, indicating a defect in osteoclast-specific function in vivo. Lack of CD13 led to exaggerated in vitro osteoclastogenesis as indicated by significantly enhanced fusion of bone marrow-derived multinucleated osteoclasts in the presence of M-CSF and RANKL, resulting in abnormally large cells containing remarkably high numbers of nuclei. Mechanistically, while expression levels of the fusion-regulatory proteins dynamin and DC-STAMP1 must be downregulated for fusion to proceed, these are aberrantly sustained at high levels even in CD13-deficient mature multi-nucleated osteoclasts. Further, the stability of fusion-promoting proteins is maintained in the absence of CD13, implicating CD13 in protein turnover mechanisms. Together, we conclude that CD13 may regulate cell-cell fusion by controlling the expression and localization of key fusion regulatory proteins that are critical for osteoclast fusion.


Subject(s)
Bone Resorption/genetics , CD13 Antigens/genetics , CD13 Antigens/metabolism , Osteoclasts/pathology , Animals , Bone Density , Bone Resorption/pathology , Cell Differentiation , Cell Fusion , Cell Line , Female , Gene Expression Regulation , Gene Knockout Techniques , Humans , Male , Mice , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Osteoclasts/metabolism , U937 Cells
7.
Cell Rep ; 34(12): 108894, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33761361

ABSTRACT

The process of hematopoiesis is subject to substantial ontogenic remodeling that is accompanied by alterations in cellular fate during both development and disease. We combine state-of-the-art mass spectrometry with extensive functional assays to gain insight into ontogeny-specific proteomic mechanisms regulating hematopoiesis. Through deep coverage of the cellular proteome of fetal and adult lympho-myeloid multipotent progenitors (LMPPs), common lymphoid progenitors (CLPs), and granulocyte-monocyte progenitors (GMPs), we establish that features traditionally attributed to adult hematopoiesis are conserved across lymphoid and myeloid lineages, whereas generic fetal features are suppressed in GMPs. We reveal molecular and functional evidence for a diminished granulocyte differentiation capacity in fetal LMPPs and GMPs relative to their adult counterparts. Our data indicate an ontogeny-specific requirement of myosin activity for myelopoiesis in LMPPs. Finally, we uncover an ontogenic shift in the monocytic differentiation capacity of GMPs, partially driven by a differential expression of Irf8 during fetal and adult life.


Subject(s)
Cell Lineage , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Proteomics , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Cell Differentiation , Fetus/cytology , Granulocytes/cytology , HEK293 Cells , Humans , Immunophenotyping , Interferon Regulatory Factors/metabolism , Kinetics , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Mice, Inbred C57BL , Monocytes/cytology , Monocytes/metabolism , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Proteome/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
8.
J Exp Med ; 218(4)2021 04 05.
Article in English | MEDLINE | ID: mdl-33635312

ABSTRACT

Hematopoietic stem cells reside in the bone marrow, where they generate the effector cells that drive immune responses. However, in response to inflammation, some hematopoietic stem and progenitor cells (HSPCs) are recruited to tissue sites and undergo extramedullary hematopoiesis. Contrasting with this paradigm, here we show residence and differentiation of HSPCs in healthy gingiva, a key oral barrier in the absence of overt inflammation. We initially defined a population of gingiva monocytes that could be locally maintained; we subsequently identified not only monocyte progenitors but also diverse HSPCs within the gingiva that could give rise to multiple myeloid lineages. Gingiva HSPCs possessed similar differentiation potentials, reconstitution capabilities, and heterogeneity to bone marrow HSPCs. However, gingival HSPCs responded differently to inflammatory insults, responding to oral but not systemic inflammation. Combined, we highlight a novel pathway of myeloid cell development at a healthy barrier, defining a gingiva-specific HSPC network that supports generation of a proportion of the innate immune cells that police this barrier.


Subject(s)
Gingiva/cytology , Gingiva/immunology , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/immunology , Animals , Bone Marrow/metabolism , Female , Hematopoiesis , Male , Mice , Mice, Inbred C57BL , Mouth Mucosa/cytology , Mouth Mucosa/immunology , RNA-Seq/methods , Single-Cell Analysis/methods
9.
Immunity ; 53(2): 319-334.e6, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32814027

ABSTRACT

Neutrophils are the most abundant peripheral immune cells and thus, are continually replenished by bone marrow-derived progenitors. Still, how newly identified neutrophil subsets fit into the bone marrow neutrophil lineage remains unclear. Here, we use mass cytometry to show that two recently defined human neutrophil progenitor populations contain a homogeneous progenitor subset we term "early neutrophil progenitors" (eNePs) (Lin-CD66b+CD117+CD71+). Surface marker- and RNA-expression analyses, together with in vitro colony formation and in vivo adoptive humanized mouse transfers, indicate that eNePs are the earliest human neutrophil progenitors. Furthermore, we identified CD71 as a marker associated with the earliest neutrophil developmental stages. Expression of CD71 marks proliferating neutrophils, which were expanded in the blood of melanoma patients and detectable in blood and tumors from lung cancer patients. In summary, we establish CD117+CD71+ eNeP as the inceptive human neutrophil progenitor and propose a refined model of the neutrophil developmental lineage in bone marrow.


Subject(s)
Antigens, CD/metabolism , Bone Marrow Cells/cytology , Myeloid Progenitor Cells/metabolism , Neutrophils/cytology , Proto-Oncogene Proteins c-kit/metabolism , Receptors, Transferrin/metabolism , Adoptive Transfer , Animals , Bone Marrow/metabolism , Cell Lineage , Humans , Male , Melanoma/blood , Mice , Mice, Inbred NOD , Myeloid Progenitor Cells/cytology
10.
Nature ; 582(7813): 571-576, 2020 06.
Article in English | MEDLINE | ID: mdl-32499656

ABSTRACT

Macrophages are the first cells of the nascent immune system to emerge during embryonic development. In mice, embryonic macrophages infiltrate developing organs, where they differentiate symbiotically into tissue-resident macrophages (TRMs)1. However, our understanding of the origins and specialization of macrophages in human embryos is limited. Here we isolated CD45+ haematopoietic cells from human embryos at Carnegie stages 11 to 23 and subjected them to transcriptomic profiling by single-cell RNA sequencing, followed by functional characterization of a population of CD45+CD34+CD44+ yolk sac-derived myeloid-biased progenitors (YSMPs) by single-cell culture. We also mapped macrophage heterogeneity across multiple anatomical sites and identified diverse subsets, including various types of embryonic TRM (in the head, liver, lung and skin). We further traced the specification trajectories of TRMs from either yolk sac-derived primitive macrophages or YSMP-derived embryonic liver monocytes using both transcriptomic and developmental staging information, with a focus on microglia. Finally, we evaluated the molecular similarities between embryonic TRMs and their adult counterparts. Our data represent a comprehensive characterization of the spatiotemporal dynamics of early macrophage development during human embryogenesis, providing a reference for future studies of the development and function of human TRMs.


Subject(s)
Macrophages/cytology , Single-Cell Analysis , Cell Lineage , Embryo, Mammalian/cytology , Head , Hematopoiesis , Humans , Leukocyte Common Antigens/metabolism , Liver/cytology , Liver/embryology , Lung/cytology , Macrophages/metabolism , Microglia/cytology , Myeloid Progenitor Cells/cytology , RNA-Seq , Skin/cytology , Spatio-Temporal Analysis , Transcriptome , Yolk Sac/cytology
11.
Sci Rep ; 10(1): 6335, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32286456

ABSTRACT

Numerous cell types modulate hematopoiesis through soluble and membrane bound molecules. Whether developing hematopoietic progenitors of a particular lineage modulate the differentiation of other hematopoietic lineages is largely unknown. Here we aimed to investigate the influence of myeloid progenitors on CD34+ cell differentiation into CD56+ innate lymphocytes. Sorted CD34+ cells cultured in the presence of stem cell factor (SCF) and FMS-like tyrosine kinase 3 ligand (FLT3L) give rise to numerous cell types, including progenitors that expressed the prolactin receptor (PRLR). These CD34+PRLR+ myeloid-lineage progenitors were derived from granulocyte monocyte precursors (GMPs) and could develop into granulocytes in the presence of granulocyte-macrophage colony-stimulating factor (GM-CSF) in vitro. Moreover, CD34+PRLR+ myeloid progenitors lacked lymphoid developmental potential, but when stimulated with prolactin (PRL) they increased the differentiation of other CD34+ cell populations into the NK lineage in a non-contact dependent manner. Both mRNA and protein analyses show that PRL increased mothers against decapentaplegic homolog 7 (SMAD7) in CD34+PRLR+ myeloid cells, which reduced the production of transforming growth factor beta 1 (TGF-ß1), a cytokine known to inhibit CD56+ cell development. Thus, we uncover an axis whereby CD34+PRLR+ GMPs inhibit CD56+ lineage development through TGF-ß1 production and PRL stimulation leads to SMAD7 activation, repression of TGF-ß1, resulting in CD56+ cell development.


Subject(s)
Hematopoietic Stem Cells/metabolism , Lymphopoiesis/genetics , Prolactin/genetics , Receptors, Prolactin/genetics , Smad7 Protein/genetics , Transforming Growth Factor beta1/genetics , Antigens, CD34/genetics , Antigens, CD34/immunology , CD56 Antigen/genetics , CD56 Antigen/immunology , Cell Differentiation/genetics , Cell Lineage/genetics , Gene Expression Regulation, Developmental/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Lymphocytes/cytology , Lymphocytes/immunology , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , fms-Like Tyrosine Kinase 3/genetics
12.
Dev Cell ; 53(2): 229-239.e7, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32197069

ABSTRACT

Natural killer (NK) cells are a critical component of the innate immune system. However, their ontogenic origin has remained unclear. Here, we report that NK cell potential first arises from Hoxaneg/low Kit+CD41+CD16/32+ hematopoietic-stem-cell (HSC)-independent erythro-myeloid progenitors (EMPs) present in the murine yolk sac. EMP-derived NK cells and primary fetal NK cells, unlike their adult counterparts, exhibit robust degranulation in response to stimulation. Parallel studies using human pluripotent stem cells (hPSCs) revealed that HOXAneg/low CD34+ progenitors give rise to NK cells that, similar to murine EMP-derived NK cells, harbor a potent cytotoxic degranulation bias. In contrast, hPSC-derived HOXA+ CD34+ progenitors, as well as human cord blood CD34+ cells, give rise to NK cells that exhibit an attenuated degranulation response but robustly produce inflammatory cytokines. Collectively, our studies identify an extra-embryonic origin of potently cytotoxic NK cells, suggesting that ontogenic origin is a relevant factor in designing hPSC-derived adoptive immunotherapies.


Subject(s)
Cell Differentiation , Cell Lineage , Embryonic Stem Cells/cytology , Erythroid Precursor Cells/cytology , Hematopoietic Stem Cells/cytology , Killer Cells, Natural/pathology , Myeloid Progenitor Cells/cytology , Animals , Embryonic Stem Cells/metabolism , Erythroid Precursor Cells/metabolism , Female , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Humans , Killer Cells, Natural/metabolism , Male , Mice , Myeloid Progenitor Cells/metabolism , Yolk Sac
13.
Sci Rep ; 10(1): 1771, 2020 02 04.
Article in English | MEDLINE | ID: mdl-32019985

ABSTRACT

Immune cells resident in adipose tissue have important functions in local and systemic metabolic homeostasis. Nevertheless, these immune cell populations remain poorly characterized in bovines. Recently, we described diverse lymphocyte subpopulations in adipose tissue of Holstein-Friesian cows. Here, we aimed at characterising myeloid cell populations present in bovine adipose tissue using multicolour flow cytometry, cell sorting and histochemistry/immunohistochemistry. Macrophages, CD14+CD11b+MHC-II+CD45+ cells, were identified in mesenteric and subcutaneous adipose tissue, though at higher proportions in the latter. Mast cells, identified as SSC-AhighCD11b-/+CD14-MHC-II-CH138A-CD45+ cells, were also observed in adipose tissue and found at higher proportions than macrophages in mesenteric adipose tissue. Neutrophils, presenting a CH138A+CD11b+ phenotype, were also detected in mesenteric and subcutaneous adipose tissue, however, at much lower frequencies than in the blood. Our gating strategy allowed identification of eosinophils in blood but not in adipose tissue although being detected by morphological analysis at low frequencies in some animals. A population not expressing CD45 and with the CH138A+ CD11b-MHC-II- phenotype, was found abundant and present at higher proportions in mesenteric than subcutaneous adipose tissue. The work reported here may be useful for further studies addressing the function of the described cells.


Subject(s)
Antigens, CD/metabolism , Mesentery/metabolism , Myeloid Cells/metabolism , Subcutaneous Fat/metabolism , Animals , Cattle , Female , Flow Cytometry , Immunohistochemistry , Mast Cells/cytology , Mast Cells/metabolism , Mesentery/cytology , Myeloid Cells/cytology , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Subcutaneous Fat/cytology
14.
Brain Behav Immun ; 84: 209-217, 2020 02.
Article in English | MEDLINE | ID: mdl-31812778

ABSTRACT

There has been a growing recognition of the involvement of the gastrointestinal microbiota in the development of stress-related disorders. Acute stress leads to activation of neuroendocrine systems, which in turn orchestrate a large-scale redistribution of innate immune cells. Both these response systems are independently known to be primed by the microbiota, even though much is still unclear about the role of the gastrointestinal microbiota in acute stress-induced immune activation. In this study, we investigated whether the microbiota influences acute stress-induced changes in innate immunity using conventionally colonised mice, mice devoid of any microbiota (i.e. germ-free, GF), and colonised GF mice (CGF). We also explored the kinetics of stress-induced immune cell mobilisation in the blood, the spleen and mesenteric lymph nodes (MLNs). Mice were either euthanised prior to stress or underwent restraint stress and were then euthanised at various time points (i.e. 0, 45- and 240-minutes) post-stress. Plasma adrenaline and noradrenaline levels were analysed using ELISA and immune cell levels were quantified using flow cytometry. GF mice had increased baseline levels of adrenaline and noradrenaline, of which adrenaline was normalised in CGF mice. In tandem, GF mice had decreased circulating levels of LY6Chi and LY6Cmid, CCR2+ monocytes, and granulocytes, but not LY6C-, CX3CR1+ monocytes. These deficits were normalised in CGF mice. Acute stress decreased blood LY6Chi and LY6Cmid, CCR2+ monocytes while increasing granulocyte levels in all groups 45 min post-stress. However, only GF mice showed stress-induced changes in LY6Chi monocytes and granulocytes 240 min post-stress, indicating impairments in the recovery from acute stress-induced changes in levels of specific innate immune cell types. LY6C-, CX3CR1+ monocytes remained unaffected by stress, indicating that acute stress impacts systemic innate immunity in a cell-type-specific manner. Overall, these data reveal novel cell-type-specific changes in the innate immune system in response to acute stress, which in turn are impacted by the microbiota. In conclusion, the microbiota influences the priming and recovery of the innate immune system to an acute stressor and may inform future microbiota-targeted therapeutics aimed at modulating stress-induced immune activation in stress-related disorders.


Subject(s)
Cell Movement , Gastrointestinal Microbiome , Host Microbial Interactions , Immunity, Innate , Monocytes , Stress, Physiological , Animals , Gastrointestinal Microbiome/immunology , Host Microbial Interactions/immunology , Mice , Mice, Inbred C57BL , Myeloid Progenitor Cells/cytology , Stress, Physiological/immunology
15.
Blood Adv ; 3(22): 3635-3649, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31751474

ABSTRACT

Serine palmitoyltransferase (SPT) long-chain base subunit 1 (SPTLC1) is 1 of the 2 main catalytic subunits of the SPT complex, which catalyzes the first and rate-limiting step of sphingolipid biosynthesis. Here, we show that Sptlc1 deletion in adult bone marrow (BM) cells results in defective myeloid differentiation. In chimeric mice from noncompetitive BM transplant assays, there was an expansion of the Lin- c-Kit+ Sca-1+ compartment due to increased multipotent progenitor production, but myeloid differentiation was severely compromised. We also show that defective biogenesis of sphingolipids in the endoplasmic reticulum (ER) leads to ER stress that affects myeloid differentiation. Furthermore, we demonstrate that transient accumulation of fatty acid, a substrate for sphingolipid biosynthesis, could be partially responsible for the ER stress. Independently, we find that ER stress in general, such as that induced by the chemical thapsigargin or the fatty acid palmitic acid, compromises myeloid differentiation in culture. These results identify perturbed sphingolipid metabolism as a source of ER stress, which may produce diverse pathological effects related to differential cell-type sensitivity.


Subject(s)
Cell Differentiation/genetics , Hematopoiesis/genetics , Homeostasis , Myeloid Cells/cytology , Myeloid Cells/metabolism , Serine C-Palmitoyltransferase/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Computational Biology/methods , Gene Deletion , Gene Expression Profiling , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Knockout , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Spleen/cytology , Spleen/metabolism
16.
Stem Cell Reports ; 13(6): 1099-1110, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31708474

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) can serve as a versatile and scalable source of neutrophils for biomedical research and transfusion therapies. Here we describe a rapid efficient serum- and xenogen-free protocol for neutrophil generation, which is based on direct hematoendothelial programming of hiPSCs using ETV2-modified mRNA. Culture of ETV2-induced hematoendothelial progenitors in the presence of GM-CSF, FGF2, and UM171 led to continuous production of generous amounts of CD34+CD33+ myeloid progenitors which could be harvested every 8-10 days for up to 30 days of culture. Subsequently, myeloid progenitors were differentiated into neutrophils in the presence of G-CSF and the retinoic acid agonist Am580. Neutrophils obtained in these conditions displayed a typical somatic neutrophil morphology, produced reactive oxygen species, formed neutrophil extracellular traps and possessed phagocytic and chemotactic activities. Overall, this technology offers an opportunity to generate a significant number of neutrophils as soon as 14 days after initiation of differentiation.


Subject(s)
Cell Differentiation/genetics , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Neutrophils/immunology , Neutrophils/metabolism , RNA, Messenger , Transcription Factors/genetics , Biomarkers , Cells, Cultured , Extracellular Traps/genetics , Extracellular Traps/metabolism , Gene Expression Regulation, Developmental , Hematopoiesis , Humans , Immunophenotyping , Leukopoiesis/genetics , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Neutrophils/cytology
17.
J Transl Med ; 17(1): 247, 2019 07 31.
Article in English | MEDLINE | ID: mdl-31366356

ABSTRACT

BACKGROUND: The molecular pathways that drive bone marrow myeloid progenitors (BMMP) development are very well understood and include a tight controlled multi-stage gene hierarch. Monocytes are versatile cells that display remarkable plasticity and may give rise to specific subsets of macrophages to proper promote tissue homesostasis upon an injury. However, the epigenetic mechanisms that underlie monocyte differentiation into the pro-inflammatory Ly6Chigh or the repairing Ly6Clow subsets are yet to be elucidated. We have previously shown that Epigenetic mechanisms Histone Deacetylase (HDAC) dependent are crucial for monocyte behavior and plasticity and in this work, we propose that this same mechanism underlies BMMP plasticity upon an inflammatory challenge in vivo. METHODS: BMMP were culture in the presence of GM-CSF alone or in combination with HDAC inhibitor (iHDAC) and phenotyped by flow cytometry, immune staining or western blot. iHDAC was topically added to skin wounds for 7 consecutive days and wound healing was monitored by flow cytometry and histopathological analysis. RESULTS: When BMMP were cultured in the presence of iHDAC, we showed that the CD11blow/Ly6Clow subset was the specific target of iHDAC that underwent chromatin hyperacetylation in vitro. Upon 13 days in the presence of iHDAC, BMMP gave rise to very elongated macrophages, that in turn, displayed a remarkable plasticity in a HDAC activity dependent fashion. HDAC-dependent cell shape was tight related to macrophage behavior and phenotype through the control of iNOS protein levels, showing that chromatin remodeling is a key component of macrophage plasticity and function. We then hypothesized that iHDAC would modulate the inflammatory response and favor tissue repair in vivo. To test this hypothesis, we topically added iHDAC to skin wounds during 7 consecutive days and followed tissue repair dynamics. In fact, iHDAC treated skin wounds presented an increase in wound closure at day 5 that was correlated to an enrichment in the CD11blow/Ly6Clow subset and in very elongated F4/80 positives macrophages in vivo, fully recapitulating the behavior previously observed in vitro. CONCLUSION: Our work provides the biological basis that connects chromatin remodeling to phenotypic plasticity, which in turn, may become a tractable therapeutic strategy in further translational studies.


Subject(s)
Epigenome , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Myeloid Progenitor Cells/cytology , Skin/drug effects , Skin/pathology , Wound Healing , Animals , Chromatin/chemistry , Epigenesis, Genetic , Histone Deacetylases/genetics , Humans , Inflammation , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Monocytes/cytology , Myeloid Progenitor Cells/drug effects , Phenotype
19.
Nat Immunol ; 20(7): 852-864, 2019 07.
Article in English | MEDLINE | ID: mdl-31213723

ABSTRACT

Dendritic cells (DC) are currently classified as conventional DCs (cDCs) and plasmacytoid DCs (pDCs). Through a combination of single-cell transcriptomic analysis, mass cytometry, in vivo fate mapping and in vitro clonal assays, here we show that, at the single-cell level, the priming of mouse hematopoietic progenitor cells toward the pDC lineage occurs at the common lymphoid progenitor stage, indicative of early divergence of the pDC and cDC lineages. We found the transcriptional signature of a pDC precursor stage, defined here, in the IL-7Rα+ common lymphoid progenitor population and identified Ly6D, IL-7Rα, CD81 and CD2 as key markers of pDC differentiation, which distinguish pDC precursors from cDC precursors. In conclusion, pDCs developed in the bone marrow from a Ly6DhiCD2hi lymphoid progenitor cell and differentiated independently of the myeloid cDC lineage.


Subject(s)
Antigens, Ly/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Flow Cytometry , GPI-Linked Proteins/metabolism , Gene Expression , Gene Expression Profiling , Mice , Transcriptome
20.
J Cell Mol Med ; 23(8): 5128-5143, 2019 08.
Article in English | MEDLINE | ID: mdl-31210415

ABSTRACT

A novel myeloid antigen presenting cell can be generated through in vitro haematopoiesis in long-term splenic stromal cocultures. The in vivo equivalent subset was recently identified as phenotypically and functionally distinct from the spleen subsets of macrophages, conventional (c) dendritic cells (DC), resident monocytes, inflammatory monocytes and eosinophils. This novel subset which is myeloid on the basis of cell surface phenotype, but dendritic-like on the basis of cell surface marker expression and antigen presenting function, has been tentatively labelled "L-DC." Transcriptome analysis has now been employed to determine the lineage relationship of this cell type with known splenic cDC and monocyte subsets. Principal components analysis showed separation of "L-DC" and monocytes from cDC subsets in the second principal component. Hierarchical clustering then indicated a close lineage relationship between this novel subset, resident monocytes and inflammatory monocytes. Resident monocytes were the most closely aligned, with no genes specifically expressed by the novel subset. This subset, however, showed upregulation of genes reflecting both dendritic and myeloid lineages, with strong upregulation of several genes, particularly CD300e. While resident monocytes were found to be dependent on Toll-like receptor signalling for development and were reduced in number in Myd88-/- and Trif-/- mutant mice, both the novel subset and inflammatory monocytes were unaffected. Here, we describe a novel myeloid cell type closely aligned with resident monocytes in terms of lineage but distinct in terms of development and functional capacity.


Subject(s)
Cell Lineage/genetics , Hematopoiesis/genetics , Myeloid Cells/cytology , Spleen/metabolism , Animals , Antigen Presentation , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/metabolism , Cell Differentiation/genetics , Coculture Techniques , Dendritic Cells/cytology , Dendritic Cells/metabolism , Eosinophils/cytology , Eosinophils/metabolism , Gene Expression Profiling , Lymphocyte Activation/genetics , Mice , Monocytes/cytology , Monocytes/metabolism , Myeloid Cells/metabolism , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Spleen/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...