Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 751
Filter
Add more filters

Publication year range
1.
Immunity ; 47(5): 890-902.e4, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29166589

ABSTRACT

Granulocyte-monocyte progenitors (GMPs) and monocyte-dendritic cell progenitors (MDPs) produce monocytes during homeostasis and in response to increased demand during infection. Both progenitor populations are thought to derive from common myeloid progenitors (CMPs), and a hierarchical relationship (CMP-GMP-MDP-monocyte) is presumed to underlie monocyte differentiation. Here, however, we demonstrate that mouse MDPs arose from CMPs independently of GMPs, and that GMPs and MDPs produced monocytes via similar but distinct monocyte-committed progenitors. GMPs and MDPs yielded classical (Ly6Chi) monocytes with gene expression signatures that were defined by their origins and impacted their function. GMPs produced a subset of "neutrophil-like" monocytes, whereas MDPs gave rise to a subset of monocytes that yielded monocyte-derived dendritic cells. GMPs and MDPs were also independently mobilized to produce specific combinations of myeloid cell types following the injection of microbial components. Thus, the balance of GMP and MDP differentiation shapes the myeloid cell repertoire during homeostasis and following infection.


Subject(s)
Dendritic Cells/physiology , Granulocyte Precursor Cells/physiology , Monocytes/physiology , Myeloid Progenitor Cells/physiology , Animals , Antigens, Ly/analysis , Cell Differentiation , Leukosialin/analysis , Mice , Sequence Analysis, RNA , Transcriptome
2.
J Virol ; 95(12)2021 05 24.
Article in English | MEDLINE | ID: mdl-33827945

ABSTRACT

Immune memory represents the most efficient defense against invasion and transmission of infectious pathogens. In contrast to memory T and B cells, the roles of innate immunity in recall responses remain inconclusive. In this study, we identified a novel mouse spleen NK cell subset expressing NKp46 and NKG2A induced by intranasal influenza virus infection. These memory NK cells specifically recognize N-linked glycosylation sites on influenza hemagglutinin (HA) protein. Different from memory-like NK cells reported previously, these NKp46+ NKG2A+ memory NK cells exhibited HA-specific silence of cytotoxicity but increase of gamma interferon (IFN-γ) response against influenza virus-infected cells, which could be reversed by pifithrin-µ, a p53-heat shock protein 70 (HSP70) signaling inhibitor. During recall responses, splenic NKp46+ NKG2A+ NK cells were recruited to infected lung and modulated viral clearance of virus and CD8+ T cell distribution, resulting in improved clinical outcomes. This long-lived NK memory bridges innate and adaptive immune memory response and promotes the homeostasis of local environment during recall response.IMPORTANCE In this study, we demonstrate a novel hemagglutinin (HA)-specific NKp46+ NKG2A+ NK cell subset induced by influenza A virus infection. These memory NK cells show virus-specific decreased cytotoxicity and increased gamma interferon (IFN-γ) on reencountering the same influenza virus antigen. In addition, they modulate host recall responses and CD8 T cell distribution, thus bridging the innate immune and adaptive immune responses during influenza virus infection.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunologic Memory , Influenza A Virus, H1N1 Subtype/immunology , Killer Cells, Natural/immunology , Orthomyxoviridae Infections/immunology , Adoptive Transfer , Animals , Antigens, Ly/analysis , Antigens, Ly/metabolism , Benzothiazoles/pharmacology , CD8-Positive T-Lymphocytes/immunology , Coculture Techniques , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Influenza A Virus, H9N2 Subtype/immunology , Interferon-gamma/metabolism , Killer Cells, Natural/drug effects , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily C/analysis , Natural Cytotoxicity Triggering Receptor 1/analysis , Natural Cytotoxicity Triggering Receptor 1/metabolism , Spleen/cytology , Spleen/immunology , Toluene/analogs & derivatives , Toluene/pharmacology
3.
Pancreatology ; 22(2): 294-303, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35120820

ABSTRACT

BACKGROUND & AIMS: Sca-1 is a surface marker for murine hematopoietic stem cells (HSCs) and type-I interferon is a key regulator for Lin-Sca-1+ HSCs expansion through Ifnar/Stat-1/Sca-1-signaling. In this study we aimed to characterize the role and regulation of Sca-1+ cells in pancreatic regeneration. METHODS: To characterize Sca-1 in vivo, immunohistochemistry and immunofluorescence staining of Sca-1 was conducted in normal pancreas, in cerulein-mediated acute pancreatitis, and in Kras-triggered cancerous lesions. Ifnar/Stat-1/Sca-1-signaling was studied in type-I IFN-treated epithelial explants of adult wildtype, Ifnar-/-, and Stat-1-/- mice. Sca-1 induction was analyzed by gene expression and FACS analysis. After isolation of pancreatic epithelial Lin-Sca-1+cells, pancreatosphere-formation and immunofluorescence-assays were carried out to investigate self-renewal and differentiation capabilities. RESULTS: Sca-1+ cells were located in periacinar and periductal spaces and showed an enrichment during cerulein-induced acute pancreatitis (23.2/100 µm2 ± 4.9 SEM) and in early inflammation-mediated carcinogenic lesions of the pancreas of KrasG12D mice (35.8/100 µm2 ± SEM 1.9) compared to controls (3.6/100 µm2 ± 1.3 SEM). Pancreatic Lin-Sca-1+ cells displayed a small population of 1.46% ± 0.12 SEM in FACS. In IFN-ß treated pancreatic epithelial explants, Sca-1 expression was increased, and Lin-Sca-1+ cells were enriched in vitro (from 1.49% ± 0.36 SEM to 3.85% ± 0.78 SEM). Lin-Sca-1+ cells showed a 12 to 51-fold higher capacity for clonal self-renewal compared to Lin-Sca-1- cells and generated cells express markers of the acinar and ductal compartment. CONCLUSIONS: Pancreatic Sca-1+ cells enriched during parenchymal damage showed a significant capacity for cell renewal and in vitro plasticity, suggesting that corresponding to the type I interferon-dependent regulation of Lin-Sca-1+ hematopoietic stem cells, pancreatic Sca-1+ cells also employ type-I-interferon for regulating progenitor-cell-homeostasis.


Subject(s)
Cell Plasticity , Pancreatitis , Acute Disease , Animals , Antigens, Ly/analysis , Antigens, Ly/genetics , Antigens, Ly/metabolism , Epithelial Cells , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Pancreas/pathology , Pancreatitis/chemically induced , Pancreatitis/genetics , Pancreatitis/pathology
4.
Immunity ; 36(6): 1047-59, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22749354

ABSTRACT

Dendritic cells (DCs), monocytes, and/or macrophages initiate host-protective immune responses to intracellular pathogens in part through interleukin-12 (IL-12) production, although the relative contribution of tissue resident versus recruited cells has been unclear. Here, we showed that after intraperitoneal infection with Toxoplasma gondii cysts, resident mononuclear phagocytes are replaced by circulating monocytes that differentiate in situ into inflammatory DCs (moDCs) and F4/80(+) macrophages. Importantly, NK cell-derived interferon-γ (IFN-γ) was required for both the loss of resident mononuclear phagocytes and the local differentiation of monocytes into macrophages and moDCs. This newly generated moDC population and not the resident DCs (or macrophages) served as the major source of IL-12 at the site of infection. Thus, NK cell-derived IFN-γ is important in both regulating inflammatory cell dynamics and in driving the local differentiation of monocytes into the cells required for initiating the immune response to an important intracellular pathogen.


Subject(s)
Dendritic Cells/immunology , Interferon-gamma/physiology , Killer Cells, Natural/immunology , Monocytes/immunology , Adoptive Transfer , Animals , Antigens, Ly/analysis , Cell Differentiation , Chemotaxis, Leukocyte , Dendritic Cells/pathology , Dendritic Cells/transplantation , Genes, Reporter , Interleukin-12 Subunit p40/biosynthesis , Interleukin-12 Subunit p40/genetics , Killer Cells, Natural/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/transplantation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/chemistry , Monocytes/pathology , Monocytes/transplantation , Myeloid Differentiation Factor 88/physiology , Neutrophils/immunology , Peritonitis/immunology , Peritonitis/parasitology , Phagocytes/classification , Phagocytes/immunology , Phagocytes/pathology , Receptors, Interferon/deficiency , Receptors, Interferon/physiology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Specific Pathogen-Free Organisms , T-Lymphocyte Subsets/immunology , Toxoplasmosis, Animal/immunology , Interferon gamma Receptor
5.
EMBO J ; 35(6): 685-98, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26884587

ABSTRACT

Monocytes have emerged as critical driving force of acute inflammation. Here, we show that inhibition of Toll-like receptor 2(TLR2) dimerization by a TLR2 transmembrane peptide (TLR2-p) ameliorated DSS-induced colitis by interfering specifically with the activation of Ly6C(+) monocytes without affecting their recruitment to the colon. We report that TLR2-p directly interacts with TLR2 within the membrane, leading to inhibition of TLR2-TLR6/1 assembly induced by natural ligands. This was associated with decreased levels of extracellular signal-regulated kinases (ERK) signaling and reduced secretion of pro-inflammatory cytokines, such as interleukin (IL)-6, IL-23, IL-12, and IL-1ß. Altogether, our study provides insights into the essential role of TLR2 dimerization in the activation of pathogenic pro-inflammatory Ly6C(hi) monocytes and suggests that inhibition of this aggregation by TLR2-p might have therapeutic potential in the treatment of acute gut inflammation.


Subject(s)
Colitis/pathology , Colon/immunology , Monocytes/drug effects , Monocytes/immunology , Protein Multimerization , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 2/metabolism , Animals , Antigens, Ly/analysis , Colitis/chemically induced , Cytokines/metabolism , Disease Models, Animal , Immunophenotyping , MAP Kinase Signaling System , Mice, Inbred C57BL , Monocytes/chemistry , Toll-Like Receptor 6/metabolism
6.
J Biol Chem ; 293(35): 13452-13463, 2018 08 31.
Article in English | MEDLINE | ID: mdl-30018139

ABSTRACT

The Lin-c-Kit+ Sca-1+ cell population in the bone marrow (BM) serves as the direct precursor for differentiation of myeloid cells. In this study, we report that deficiency in Fpr2, a G protein-coupled chemoattractant receptor in mice, is associated with reduced BM nucleated cells, including CD31+Ly6C+ (granulocytes and monocytes), CD31-/Ly6Cint (granuloid cells), and CD31-/Ly6Chigh (predominantly monocytes) cells. In particular, the number of Lin-c-Kit+Sca-1+ (LKS) cells was reduced in Fpr2-/- mouse BM. This was supported by observations of the reduced incorporation of intraperitoneally injected bromodeoxyuridine by cells in the c-Kit+ population from Fpr2-/- mouse BM. Purified c-Kit+ cells from Fpr2-/- mice showed reduced expansion when cultured in vitro with stem cell factor (SCF). SCF/c-Kit-mediated phosphorylation of P38, STAT1, Akt (Thr-308), and Akt (Ser-473) was also significantly reduced in c-Kit+ cells from Fpr2-/- mice. Furthermore, Fpr2 agonists enhanced SCF-induced proliferation of c-Kit+ cells. Colony-forming unit assays revealed that CFU-granulocyte-macrophage formation of BM cells from Fpr2-/- mice was significantly reduced. After heat-inactivated bacterial stimulation in the airway, the expansion of c-kit+ Sca-1+ cells in BM and recruitment of Ly6G+ cells to the lungs and CD11b+Ly6C+TNFα+ cells to the spleen of Fpr2-/- mice was significantly reduced. These results demonstrate an important role for Fpr2 in the development of myeloid lineage precursors in mouse BM.


Subject(s)
Antigens, Ly/analysis , Gene Deletion , Membrane Proteins/analysis , Myeloid Progenitor Cells/cytology , Proto-Oncogene Proteins c-kit/analysis , Receptors, Formyl Peptide/genetics , Animals , Cell Count , Cell Lineage , Cell Proliferation , Female , Male , Mice , Myeloid Progenitor Cells/metabolism , Receptors, Formyl Peptide/analysis
7.
J Immunol ; 199(4): 1362-1371, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28674182

ABSTRACT

Enteric pathogens including Salmonella enteric serovar Typhimurium can breach the epithelial barrier of the host and spread to systemic tissues. In response to infection, the host activates innate immune receptors via the signaling molecule MyD88, which induces protective inflammatory and antimicrobial responses. Most of these innate immune responses have been studied in hematopoietic cells, but the role of MyD88 signaling in other cell types remains poorly understood. Surprisingly, we found that Dermo1-Cre;Myd88fl/fl mice with mesenchymal cell-specific deficiency of MyD88 were less susceptible to orogastric and i.p. STyphimurium infection than their Myd88fl/fl littermates. The reduced susceptibility of Dermo1-Cre;Myd88fl/fl mice to infection was associated with lower loads of S. Typhimurium in the liver and spleen. Mutant analyses revealed that S. Typhimurium employs its virulence type III secretion system 2 to promote its growth through MyD88 signaling pathways in mesenchymal cells. Inflammatory monocytes function as a major cell population for systemic dissemination of S. Typhimurium Mechanistically, mesenchymal cell-specific MyD88 signaling promoted CCL2 production in the liver and spleen and recruitment of inflammatory monocytes to systemic organs in response to STyphimurium infection. Consistently, MyD88 signaling in mesenchymal cells enhanced the number of phagocytes including Ly6ChiLy6G- inflammatory monocytes harboring STyphimurium in the liver. These results suggest that S. Typhimurium promotes its systemic growth and dissemination through MyD88 signaling pathways in mesenchymal cells.


Subject(s)
Monocytes/immunology , Monocytes/microbiology , Myeloid Differentiation Factor 88/metabolism , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/pathogenicity , Animals , Antigens, Ly/analysis , Bacterial Load , Chemokine CCL2/biosynthesis , Immunity, Innate , Liver/immunology , Liver/microbiology , Liver/pathology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Salmonella typhimurium/immunology , Salmonella typhimurium/metabolism , Signal Transduction , Spleen/immunology , Spleen/microbiology , Type III Secretion Systems/metabolism
8.
J Immunol ; 199(4): 1261-1274, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28696256

ABSTRACT

Dead cells accumulating in the tissues may contribute to chronic inflammation. We examined the cause of impaired apoptotic cell clearance in human and murine lupus. Dead cells accumulated in bone marrow from lupus patients but not from nonautoimmune patients undergoing myeloablation, where they were efficiently removed by macrophages (MΦ). Impaired apoptotic cell uptake by MΦ also was seen in mice treated i.p. with pristane (develop lupus) but not mineral oil (MO) (do not develop lupus). The inflammatory response to both pristane and MO rapidly depleted resident (Tim4+) large peritoneal MΦ. The peritoneal exudate of pristane-treated mice contained mainly Ly6Chi inflammatory monocytes; whereas in MO-treated mice, it consisted predominantly of a novel subset of highly phagocytic MΦ resembling small peritoneal MΦ (SPM) that expressed CD138+ and the scavenger receptor Marco. Treatment with anti-Marco-neutralizing Abs and the class A scavenger receptor antagonist polyinosinic acid inhibited phagocytosis of apoptotic cells by CD138+ MΦ. CD138+ MΦ expressed IL-10R, CD206, and CCR2 but little TNF-α or CX3CR1. They also expressed high levels of activated CREB, a transcription factor implicated in generating alternatively activated MΦ. Similar cells were identified in the spleen and lung of MO-treated mice and also were induced by LPS. We conclude that highly phagocytic, CD138+ SPM-like cells with an anti-inflammatory phenotype may promote the resolution of inflammation in lupus and infectious diseases. These SPM-like cells are not restricted to the peritoneum and may help clear apoptotic cells from tissues such as the lung, helping to prevent chronic inflammation.


Subject(s)
Lupus Erythematosus, Systemic/immunology , Macrophages, Peritoneal/immunology , Phagocytosis , Syndecan-1/immunology , Animals , Antigens, Ly/analysis , Apoptosis , Bone Marrow Cells/immunology , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Humans , Inflammation/immunology , Interleukin-10 Receptor alpha Subunit/genetics , Interleukin-10 Receptor alpha Subunit/immunology , Lung/cytology , Lung/immunology , Lupus Erythematosus, Systemic/chemically induced , Lupus Erythematosus, Systemic/physiopathology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mineral Oil/pharmacology , Poly I/pharmacology , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Spleen/cytology , Spleen/immunology , Syndecan-1/genetics , Terpenes/pharmacology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
9.
Proc Natl Acad Sci U S A ; 113(33): E4847-56, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27482114

ABSTRACT

Inflammation is characterized by the recruitment of leukocytes from the bloodstream. The rapid arrival of neutrophils is followed by a wave of inflammatory lymphocyte antigen 6 complex (Ly6C)-positive monocytes. In contrast Ly6C(low) monocytes survey the endothelium in the steady state, but their role in inflammation is still unclear. Here, using confocal intravital microscopy, we show that upon Toll-like receptor 7/8 (TLR7/8)-mediated inflammation of mesenteric veins, platelet activation drives the rapid mobilization of Ly6C(low) monocytes to the luminal side of the endothelium. After repeatedly interacting with platelets, Ly6C(low) monocytes commit to a meticulous patrolling of the endothelial wall and orchestrate the subsequent arrival and extravasation of neutrophils through the production of proinflammatory cytokines and chemokines. At a molecular level, we show that cysteine-rich protein 61 (CYR61)/CYR61 connective tissue growth factor nephroblastoma overexpressed 1 (CCN1) protein is released by activated platelets and enables the recruitment of Ly6C(low) monocytes upon vascular inflammation. In addition endothelium-bound CCN1 sustains the adequate patrolling of Ly6C(low) monocytes both in the steady state and under inflammatory conditions. Blocking CCN1 or platelets with specific antibodies impaired the early arrival of Ly6C(low) monocytes and abolished the recruitment of neutrophils. These results refine the leukocyte recruitment cascade model by introducing endothelium-bound CCN1 as an inflammation mediator and by demonstrating a role for platelets and patrolling Ly6C(low) monocytes in acute vascular inflammation.


Subject(s)
Antigens, Ly/analysis , Cysteine-Rich Protein 61/physiology , Monocytes/physiology , Vasculitis/etiology , Animals , Blood Platelets/physiology , Cell Movement , Mice , Mice, Inbred C57BL , Neutrophils/physiology , Toll-Like Receptor 7/physiology , Toll-Like Receptor 8/physiology
10.
Int J Mol Sci ; 20(8)2019 Apr 16.
Article in English | MEDLINE | ID: mdl-30995786

ABSTRACT

Mouse activating Nkrp1 proteins are commonly described as type II transmembrane receptors with disulfide-linked homodimeric structure. Their function and the manner in which Nkrp1 proteins of mouse strain (C57BL/6) oligomerize are still poorly understood. To assess the oligomerization state of Nkrp1 proteins, mouse activating EGFP-Nkrp1s were expressed in mammalian lymphoid cells and their oligomerization evaluated by Förster resonance energy transfer (FRET). Alternatively, Nkrp1s oligomers were detected by Western blotting to specify the ratio between monomeric and dimeric forms. We also performed structural characterization of recombinant ectodomains of activating Nkrp1 receptors. Nkrp1 isoforms c1, c2 and f were expressed prevalently as homodimers, whereas the Nkrp1a displays larger proportion of monomers on the cell surface. Cysteine-to-serine mutants revealed the importance of all stalk cysteines for protein dimerization in living cells with a major influence of cysteine at position 74 in two Nkrp1 protein isoforms. Our results represent a new insight into the oligomerization of Nkrp1 receptors on lymphoid cells, which will help to determine their function.


Subject(s)
Antigens, Ly/analysis , NK Cell Lectin-Like Receptor Subfamily B/analysis , Receptors, Immunologic/analysis , Animals , COS Cells , Chlorocebus aethiops , Fluorescence Resonance Energy Transfer , Humans , Jurkat Cells , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily B/chemistry , Protein Multimerization , Protein Refolding
11.
Eur J Immunol ; 47(8): 1386-1388, 2017 08.
Article in English | MEDLINE | ID: mdl-28654161

ABSTRACT

Plasma cells are rare cells that have been notoriously difficult to detect by flow cytometry. New advances have described B220+ CD138+ plasma cells in the bone marrow that are particularly difficult to distinguish between CD138 intermediate B220+ developing B cells. Herein we describe a novel method for detecting plasma cells in the bone marrow using a combination of CD138 and Sca-1 staining.


Subject(s)
Antigens, Ly/analysis , Flow Cytometry/methods , Immunophenotyping/methods , Membrane Proteins/analysis , Plasma Cells/classification , Plasma Cells/immunology , Syndecan-1/analysis , Animals , Bone Marrow Cells/immunology , Leukocyte Common Antigens/analysis , Mice , Peyer's Patches/cytology , Peyer's Patches/immunology , Positive Regulatory Domain I-Binding Factor 1 , Spleen/cytology , Spleen/immunology , Transcription Factors/analysis
12.
Proc Natl Acad Sci U S A ; 112(49): E6808-17, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26598697

ABSTRACT

Pain hypersensitivity at the site of inflammation as a result of chronic immune diseases, pathogenic infection, and tissue injury is a common medical condition. However, the specific contributions of the innate and adaptive immune system to the generation of pain during inflammation have not been systematically elucidated. We therefore set out to characterize the cellular and molecular immune response in two widely used preclinical models of inflammatory pain: (i) intraplantar injection of complete Freund's adjuvant (CFA) as a model of adjuvant- and pathogen-based inflammation and (ii) a plantar incisional wound as a model of tissue injury-based inflammation. Our findings reveal differences in temporal patterns of immune cell recruitment and activation states, cytokine production, and pain in these two models, with CFA causing a nonresolving granulomatous inflammatory response whereas tissue incision induced resolving immune and pain responses. These findings highlight the significant differences and potential clinical relevance of the incisional wound model compared with the CFA model. By using various cell-depletion strategies, we find that, whereas lymphocyte antigen 6 complex locus G (Ly)6G(+)CD11b(+) neutrophils and T-cell receptor (TCR) ß(+) T cells do not contribute to the development of thermal or mechanical pain hypersensitivity in either model, proliferating CD11b(+)Ly6G(-) myeloid cells were necessary for mechanical hypersensitivity during incisional pain, and, to a lesser extent, CFA-induced inflammation. However, inflammatory (CCR2(+)Ly6C(hi)) monocytes were not responsible for these effects. The finding that a population of proliferating CD11b(+)Ly6G(-) myeloid cells contribute to mechanical inflammatory pain provides a potential cellular target for its treatment in wound inflammation.


Subject(s)
Antigens, Ly/analysis , CD11b Antigen/analysis , Inflammation/physiopathology , Myeloid Cells/immunology , Pain/physiopathology , Animals , Chemokines/biosynthesis , Cytokines/biosynthesis , Freund's Adjuvant/pharmacology , Male , Mice , Mice, Inbred C57BL , Neutrophils/immunology
13.
Dev Biol ; 416(1): 34-41, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27235813

ABSTRACT

Hematopoietic cell generation in the midgestation mouse embryo occurs through the natural transdifferentiation of temporally and spatially restricted set of hemogenic endothelial cells. These cells take on hematopoietic fate in the aorta, vitelline and umbilical arteries and appear as hematopoietic cell clusters that emerge from the vascular wall. Genetic and live imaging data have supported this. Recently, the embryonic head has been shown to contain fully functional hematopoietic stem cells (HSC). By lineage tracing, cerebrovascular specific endothelial cells were shown to contribute to the postnatal mouse hematopoietic system. Since Ly6aGFP is a marker of all HSCs, some hematopoietic cluster cells and hemogenic endothelial cells in the midgestation mouse aorta, we examine here whether embryonic head HSCs and vascular endothelial cells are positive for this marker. Whereas some head vasculature, single hematopoietic cells and all HSCs are Ly6aGFP expressing, we do not find clusters of hematopoietic cells emerging from the cerebrovasculature that are characteristic of endothelial-to-hematopoietic transition.


Subject(s)
Antigens, Ly/analysis , Head/embryology , Membrane Proteins/analysis , Animals , Antigens, Differentiation/analysis , Female , Green Fluorescent Proteins , Hematopoietic Stem Cells , Mice , Mice, Inbred C57BL , Mice, Transgenic
14.
Dev Biol ; 415(1): 111-121, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27105579

ABSTRACT

The de novo generation of hematopoietic cells occurs during midgestation when a population of endothelial cells called hemogenic endothelium transitions into hematopoietic progenitors and stem cells. In mammalian embryos, the newly formed hematopoietic cells form clusters in the lumens of the major arteries in the embryo proper and in the vascular plexus of the yolk sac. Small clusters of hematopoietic cells that are independent of the vasculature (referred to here as extravascular islands) were shown to form in the mesentery during vascular remodeling of the vitelline artery. Using three-dimensional imaging of whole mouse embryos we demonstrate that extravascular budding of hematopoietic clusters is a more widespread phenomenon that occurs from the vitelline and the umbilical arteries both proximal to the embryo proper and distal in the extraembryonic yolk sac and placenta. Furthermore, we show that there are several mechanisms by which hematopoietic clusters leave the arteries, including vascular remodeling and extrusion. Lastly, we provide static images suggesting that extravascular islands contribute to the formation of new blood vessels. Thus, extravascular islands may represent a novel mechanism of vasculogenesis whereby established vessels contribute endothelial and hematopoietic cells to developing vascular beds.


Subject(s)
Hematopoietic Stem Cells/cytology , Mesentery/embryology , Neovascularization, Physiologic/physiology , Animals , Antigens, Ly/analysis , Core Binding Factor Alpha 2 Subunit/analysis , Lymphatic System/embryology , Membrane Proteins/analysis , Mesentery/cytology , Mice , Microscopy, Confocal , Organ Specificity , Umbilical Arteries/embryology , Vascular Remodeling , Yolk Sac/blood supply
15.
Circ Res ; 117(3): 244-53, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-25991812

ABSTRACT

RATIONALE: Proinflammatory adaptive immune responses are recognized as major drivers of atherosclerotic lesion formation. Although CD8(+) T cells have recently been proposed as a proatherogenic cell subset, their full scope of actions remains to be elucidated. OBJECTIVE: We here addressed the contribution of CD8(+) T cells to monocyte trafficking in atherosclerosis. METHOD AND RESULTS: We observed that CD8(+) T cells express proinflammatory cytokines (interferon-γ, tumor necrosis factor-α, and interleukin-12) within atherosclerotic lesions and spleens of high-fat diet-fed low-density lipoprotein receptor-deficient (Ldlr(-/-)) mice. Antibody-mediated CD8(+) T-cell depletion in high-fat diet-fed Ldlr(-/-) mice decreased atherosclerotic plaque formation, associated with decreased macrophage accumulation within lesions. Despite a reduction in vascular chemokine (CC-motif) ligand 2 and chemokine (CXC-motif) ligand 1 expression, CD8(+) T-cell depletion did not directly affect monocyte recruitment to inflamed vessels. However, CD8(+) T-cell depletion decreased chemokine (CC-motif) ligand serum concentrations and circulating Ly6C(high) monocyte counts. We further evidenced that CD8(+) T-cell depletion decreased levels of mature monocytes and myeloid granulocyte-monocyte progenitors in the bone marrow and spleen of hypercholesterolemic mice, effects that were partially reproduced by interferon-γ neutralization, showing a role for interferon-γ. CONCLUSIONS: These data suggest that CD8(+) T cells promote atherosclerosis by controlling monopoiesis and circulating monocyte levels, which ultimately contributes to plaque macrophage burden without affecting direct monocyte recruitment, identifying this cell subset as a critical regulator of proatherogenic innate immune cell responses in atherosclerosis.


Subject(s)
Antigens, Ly/analysis , Atherosclerosis/immunology , CD8-Positive T-Lymphocytes/immunology , Monocytes/immunology , Myelopoiesis/immunology , Animals , Antilymphocyte Serum/therapeutic use , Atherosclerosis/etiology , Bone Marrow/metabolism , Bone Marrow/pathology , CD8-Positive T-Lymphocytes/metabolism , Carotid Stenosis/immunology , Carotid Stenosis/pathology , Cells, Cultured , Chemotaxis, Leukocyte/immunology , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/metabolism , Diet, Atherogenic/adverse effects , Dietary Fats/toxicity , Endarterectomy, Carotid , Gene Expression Regulation/immunology , Humans , Hypercholesterolemia/complications , Hypercholesterolemia/etiology , Interferon-gamma/physiology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL/deficiency , Receptors, LDL/genetics
16.
J Am Soc Nephrol ; 27(3): 792-803, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26160897

ABSTRACT

Monocytes have a crucial role in both proinflammatory and anti-inflammatory phenomena occurring during sepsis. Monocyte recruitment and activation are orchestrated by the chemokine receptors CX3CR1 and CCR2 and their cognate ligands. However, little is known about the roles of these cells and chemokines during the acute phase of inflammation in sepsis. Using intravital microscopy in a murine model of polymicrobial sepsis, we showed that inflammatory Ly6C(high) monocytes infiltrated kidneys, exhibited altered motility, and adhered strongly to the renal vascular wall in a chemokine receptor CX3CR1-dependent manner. Adoptive transfer of Cx3cr1-proficient monocyte-enriched bone marrow cells into septic Cx3cr1-depleted mice prevented kidney damage and promoted mouse survival. Modulation of CX3CR1 activation in septic mice controlled monocyte adhesion, regulated proinflammatory and anti-inflammatory cytokine expression, and was associated with the extent of kidney lesions such that the number of lesions decreased when CX3CR1 activity increased. Consistent with these results, the pro-adhesive I249 CX3CR1 allele in humans was associated with a lower incidence of AKI in patients with sepsis. These data show that inflammatory monocytes have a protective effect during sepsis via a CX3CR1-dependent adhesion mechanism. This receptor might be a new therapeutic target for kidney injury during sepsis.


Subject(s)
Acute Kidney Injury/prevention & control , Acute-Phase Reaction/immunology , Cell Adhesion , Monocytes/transplantation , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Sepsis/complications , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Adoptive Transfer , Alleles , Animals , Antigens, Ly/analysis , CX3C Chemokine Receptor 1 , Cell Adhesion/genetics , Cell Movement , Endothelium, Vascular/metabolism , Genotype , Humans , Intravital Microscopy , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence, Multiphoton , Monocytes/chemistry , Monocytes/physiology , Polymorphism, Genetic , Receptors, Interleukin-1/antagonists & inhibitors
17.
Int J Mol Sci ; 18(10)2017 Sep 29.
Article in English | MEDLINE | ID: mdl-28961183

ABSTRACT

Macrophages predominate the inflammatory landscape within multiple sclerosis (MS) lesions, not only regarding cellularity but also with respect to the diverse functions this cell fraction provides during disease progression and remission. Researchers have been well aware of the fact that the macrophage pool during central nervous system (CNS) autoimmunity consists of a mixture of myeloid cells. Yet, separating these populations to define their unique contribution to disease pathology has long been challenging due to their similar marker expression. Sophisticated lineage tracing approaches as well as comprehensive transcriptome analysis have elevated our insight into macrophage biology to a new level enabling scientists to dissect the roles of resident (microglia and non-parenchymal macrophages) and infiltrating macrophages with unprecedented precision. To do so in an accurate way, researchers have to know their toolbox, which has been filled with diverse, discriminating approaches from decades of studying neuroinflammation in animal models. Every method has its own strengths and weaknesses, which will be addressed in this review. The focus will be on tools to manipulate and/or identify different macrophage subgroups within the injured murine CNS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Macrophages/pathology , Microglia/pathology , Animals , Antigens, Ly/analysis , CX3C Chemokine Receptor 1/analysis , Humans , Inflammation/pathology , Leukocyte Common Antigens/analysis , Receptors, CCR2/analysis
18.
J Hepatol ; 64(4): 881-90, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26639397

ABSTRACT

BACKGROUND & AIMS: The leukocyte composition of tumors is heterogeneous, as is the involvement of each leukocyte subset in promoting or restraining tumorigenesis. This heterogeneity reflects the tissue of origin, tumor stage, and the functional state of leukocyte activation, but its biological roots remain poorly understood. Since tumorigenesis is driven by various genetic events, we assessed the role of driver genes in shaping the profiles and the roles of leukocytes in tumorigenesis. METHODS: Mouse liver tumors were induced by hepatic overexpression of either MYC or the combination of myristoylated AKT and NRAS(V12) oncogenes via hydrodynamic transfection. A comparative, flow cytometry- and histology-based immunophenotyping of liver-infiltrating leukocytes was performed at various stages of liver tumorigenesis. The roles of the most abundant leukocyte subsets in tumorigenesis were addressed by immunodepletion. The contribution of liver injury was assessed by comparing the injury-inducing hydrodynamic transfection model to a model in which MYC is an inducible transgene. RESULTS: Myristoylated AKT and NRAS(V12) promoted a marked recruitment of CD11b(+)Ly6G(hi)Ly6C(int) neutrophils and CD11b(+)Ly6G(-)Ly6C(hi) monocytes to the liver, but their immunodepletion did not alter tumorigenesis. In contrast, despite minimal invasion by monocytes/neutrophils during MYC-driven tumorigenesis, immunodepletion of these cells reduced MYC tumor burden and extended survival. MYC-driven tumor initiation was augmented specifically by Ly6C+ monocytes and their ability to promote liver injury. CONCLUSIONS: Our results demonstrate that leukocyte profiles do not necessarily predict their involvement in tumorigenesis, the functional role of leukocytes can be shaped by oncogenes, and that monocyte-dependent tissue injury selectively cooperates with MYC during tumorigenesis.


Subject(s)
Genes, myc/physiology , Liver Neoplasms, Experimental/etiology , Monocytes/physiology , Animals , Antigens, Ly/analysis , Female , Genes, ras , Mice , Neutrophil Infiltration , Proto-Oncogene Proteins c-akt/genetics , Receptors, Chemokine/analysis
19.
J Hepatol ; 64(1): 128-34, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26341825

ABSTRACT

BACKGROUND & AIMS: NKp46(+) cells are major effector cells in the pathogenesis of hepatic ischemia reperfusion injury (IRI). Nevertheless, the precise role of unconventional subsets like the IL-22-producing NKp46(+) cells (NK22) remains unknown. The purpose of this study was to examine the role of NK22 cells in IRI in transplantation, particularly with respect to regulation by the transcription factor ROR-gamma-t (RORγt). METHODS: To explore the role of NK22 cells in IRI in the absence of adaptive immunity, B6.RORγt-(gfp/wt)-reporter and B6.RORγt-(gfp/gfp)-knockout (KO) mice on a Rag KO background underwent 90min partial warm ischemia, followed by 24h of reperfusion. RESULTS: Rag KO mice that possess fully functional NKp46(+) cells, and Rag-common-γ-chain-double-KO (Rag-γc-DKO) mice that lack T, B and NKp46(+) cells, were used as controls. We found that Rag-γc-DKO mice lacking NK22 cells show more severe levels of hepatocellular damage (GPT, histological injury) when compared to both Rag-RORγt-reporter and Rag KO mice that possess NK22 cells. Importantly, Rag-RORγt-reporter and Rag KO mice undergoing IRI expressed high protein levels of both IL-22 and GFP (RORγt), suggesting a protective role for RORγt(+) NK22 cells in IRI. Therefore, we tested the hypothesis that RORγt critically protects from IRI through the induction of hepatic NK22 cells by studying Rag-Rorγt-DKO mice under IRI conditions. We found that the lack of RORγt(+) NK22 cells in Rag-Rorγt-DKO mice significantly enhanced IR-induced hepatocellular injury, a phenotype that could be reversed upon adoptive transfer of Rag-Rorγt-reporter NK22 cells into DKO mice. CONCLUSIONS: RORγt(+) NK22 cells play an important protective role in IRI in mice.


Subject(s)
Antigens, Ly/physiology , Interleukins/biosynthesis , Liver/blood supply , Natural Cytotoxicity Triggering Receptor 1/physiology , Nuclear Receptor Subfamily 1, Group F, Member 3/physiology , Reperfusion Injury/prevention & control , Animals , Antigens, Ly/analysis , Homeodomain Proteins/physiology , Interferon-gamma/biosynthesis , Killer Cells, Natural/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Cytotoxicity Triggering Receptor 1/analysis , Nuclear Receptor Subfamily 1, Group F, Member 3/analysis , Reperfusion Injury/immunology , Interleukin-22
20.
J Am Soc Nephrol ; 26(9): 2183-97, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25568177

ABSTRACT

The mechanism by which glucocorticoids alleviate renal inflammatory disorders remains incompletely understood. Here, we report that the efficacy of glucocorticoids in ameliorating FSGS depends on the capacity to expand myeloid-derived suppressor cells (MDSCs). After glucocorticoid treatment, the frequency of CD11b(+)HLA-DR(-)CD14(-)CD15(+) MDSCs in peripheral blood rapidly increased in patients with glucocorticoid-sensitive FSGS but remained unchanged in patients with glucocorticoid-resistant FSGS. The frequency of CD11b(+)Gr-1(+) MDSCs in mouse peripheral blood, bone marrow, spleen, kidney-draining lymph nodes (KDLNs), and kidney also increased after glucocorticoid treatment. The induced MDSCs from glucocorticoid-treated mice strongly suppressed T cells, dendritic cells, and macrophages but induced regulatory T cells in spleen, KDLNs, and kidney. Moreover, glucocorticoid treatment suppressed doxorubicin-induced T cell proliferation, dendritic cell and macrophage infiltration, and proinflammatory cytokine production, whereas this protective effect was largely abolished by depleting MDSCs using anti-Gr-1 antibody. Finally, the adoptive transfer of induced MDSCs into the doxorubicin-treated mice not only confirmed the protective role of MDSCs in doxorubicin-induced renal injury but also showed that the transferred MDSCs rapidly migrated into the lymphocyte-accumulating organs, such as the spleen and KDLNs, where they suppressed T cell proliferation. Taken together, these results demonstrate that glucocorticoid treatment ameliorates FSGS by expanding functional MDSCs and that this rapid elevation of MDSCs in peripheral blood may serve as an indicator for predicting the efficacy of glucocorticoid treatment.


Subject(s)
Dendritic Cells/drug effects , Glomerulosclerosis, Focal Segmental/blood , Glomerulosclerosis, Focal Segmental/drug therapy , Glucocorticoids/pharmacology , Myeloid Cells/drug effects , T-Lymphocytes, Regulatory/drug effects , Adolescent , Adoptive Transfer , Adult , Animals , Antigens, Ly/analysis , CD11b Antigen/analysis , Cell Count , Cell Movement/drug effects , Cell Proliferation/drug effects , Doxorubicin/pharmacology , Female , Glomerulosclerosis, Focal Segmental/pathology , HLA-DR Antigens/analysis , Humans , Kidney/pathology , Lewis X Antigen/analysis , Lipopolysaccharide Receptors/analysis , Lymph Nodes/pathology , Macrophages/drug effects , Male , Mice , Middle Aged , Myeloid Cells/chemistry , Receptors, Chemokine/analysis , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL