Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Nat Immunol ; 23(5): 679-691, 2022 05.
Article in English | MEDLINE | ID: mdl-35484408

ABSTRACT

Here we report the identification of human CD66b-CD64dimCD115- neutrophil-committed progenitor cells (NCPs) within the SSCloCD45dimCD34+ and CD34dim/- subsets in the bone marrow. NCPs were either CD45RA+ or CD45RA-, and in vitro experiments showed that CD45RA acquisition was not mandatory for their maturation process. NCPs exclusively generated human CD66b+ neutrophils in both in vitro differentiation and in vivo adoptive transfer experiments. Single-cell RNA-sequencing analysis indicated NCPs fell into four clusters, characterized by different maturation stages and distributed along two differentiation routes. One of the clusters was characterized by an interferon-stimulated gene signature, consistent with the reported expansion of peripheral mature neutrophil subsets that express interferon-stimulated genes in diseased individuals. Finally, comparison of transcriptomic and phenotypic profiles indicated NCPs represented earlier neutrophil precursors than the previously described early neutrophil progenitors (eNePs), proNeus and COVID-19 proNeus. Altogether, our data shed light on the very early phases of neutrophil ontogeny.


Subject(s)
Antigens, CD , Bone Marrow , Cell Adhesion Molecules , Cell Differentiation , Neutrophils , Receptor, Macrophage Colony-Stimulating Factor , Receptors, IgG , Bone Marrow Cells , COVID-19 , GPI-Linked Proteins , Humans , Interferons , Neutrophils/cytology
2.
Immunol Rev ; 314(1): 111-124, 2023 03.
Article in English | MEDLINE | ID: mdl-36484356

ABSTRACT

Polymorphonuclear neutrophils are no longer considered as a homogeneous population of terminally differentiated and short-lived cells that belong to the innate immune system only. In fact, data from the past decades have uncovered that neutrophils exhibit large phenotypic heterogeneity and functional versatility that render them more plastic than previously thought. Hence, their precise role as effector cells in inflammation, in immune response and in other pathophysiological processes, including tumors, needs to be better evaluated. In such a complex scenario, common knowledge of the differentiation of neutrophils in bone marrow refers to lineage precursors, starting from the still poorly defined myeloblasts, and proceeding sequentially to promyelocytes, myelocytes, metamyelocytes, band cells, segmented neutrophils, and mature neutrophils, with each progenitor stage being more mature and better characterized. Thanks to the development and utilization of cutting-edge technologies, novel information about neutrophil precursors at stages earlier than the promyelocytes, hence closer to the hematopoietic stem cells, is emerging. Accordingly, this review discusses the main findings related to the very early precursors of human neutrophils and provides our perspectives on human neutropoiesis.


Subject(s)
Bone Marrow , Neutrophils , Humans , Hematopoietic Stem Cells , Bone Marrow Cells
3.
Pediatr Blood Cancer ; 70(12): e30671, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37712719

ABSTRACT

We report a novel case of SMARCD2 (SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily D, member 2) mutation successfully treated with hematopoietic stem cell transplantation. The female patient presented delayed cord separation, chronic diarrhea, skin abscesses, skeletal dysmorphisms, and neutropenia with specific granule deficiency. Analysis of the transcriptomic profile of peripheral blood sorted mature and immature SMARCD2 neutrophils showed defective maturation process that associated with altered expression of genes related to specific, azurophilic, and gelatinase granules, such as LTF, CRISP3, PTX3, and CHI3L1. These abnormalities account for the prevalence of immature neutrophils in the peripheral blood, impaired function, and deregulated inflammatory responses.

4.
Int J Mol Sci ; 24(3)2023 Jan 22.
Article in English | MEDLINE | ID: mdl-36768547

ABSTRACT

Apolipoprotein CIII (ApoCIII) represents a key regulator of plasma lipid metabolism and a recognized risk factor for atherosclerosis and cardiovascular diseases. Beyond the regulation of lipoprotein trafficking, ApoCIII is also involved in endothelial dysfunction and monocyte recruitment related to atherothrombosis. With tissue factor (TF) being the primary initiator of the blood coagulation cascade, we hypothesized that ApoCIII-treated monocytes could express it. Hence, human CD14+-monocytes and autologous neutrophils were incubated with ApoCIII and sera from human subjects containing previously measured ApoCIII amounts. By RT-qPCR and ELISA, CD14+-monocytes, but not neutrophils, were found to show increased mRNA expression and production of TNFα, IL-1ß and IL-6 as well as TF mRNA once exposed to ultra-purified ApoCIII. By flow cytometry, CD14+-monocytes were found to rapidly express TF on their cell surface membrane when incubated with either ApoCIII or sera with known concentrations of ApoCIII. Finally, preincubation with specific ApoCIII-neutralizing antibodies significantly reduced the ability of most sera with known concentrations of ApoCIII to upregulate TF protein, other than partially inhibiting cytokine release, in CD14+-monocytes. In sum, herein we demonstrate that ApoCIII activates CD14+-monocytes to express TF. The data identify a potential mechanism which links circulating apolipoproteins with inflammation and atherothrombosis-related processes underlying cardiovascular risk.


Subject(s)
Monocytes , Thromboplastin , Humans , Apolipoprotein C-III/metabolism , Apolipoproteins/genetics , Apolipoproteins/metabolism , Monocytes/metabolism , RNA, Messenger/metabolism , Thromboplastin/genetics , Thromboplastin/metabolism
5.
FASEB J ; 34(7): 9269-9284, 2020 07.
Article in English | MEDLINE | ID: mdl-32413173

ABSTRACT

Monocytic cells perform crucial homeostatic and defensive functions. However, their fate and characterization at the transcriptomic level in human tissues are partially understood, often as a consequence of the lack of specific markers allowing their unequivocal identification. The 6-sulfo LacNAc (slan) antigen identifies a subset of non-classical (NC) monocytes in the bloodstream, namely the slan+ -monocytes. In recent studies, we and other groups have reported that, in tonsils, slan marks dendritic cell (DC)-like cells, as defined by morphological, phenotypical, and functional criteria. However, subsequent investigations in lymphomas have uncovered a significant heterogeneity of tumor-infiltrating slan+ -cells, including a macrophage-like state. Based on their emerging role in tissue inflammation and cancer, herein we investigated slan+ -cell fate in tonsils by using a molecular-based approach. Hence, RNA from tonsil slan+ -cells, conventional CD1c+ DCs (cDC2) and CD11b+ CD14+ -macrophages was subjected to gene expression analysis. For comparison, transcriptomes were also obtained from blood cDC2, classical (CL), intermediate (INT), NC, and slan+ -monocytes. Data demonstrate that the main trajectory of human slan+ -monocytes infiltrating the tonsil tissue is toward a macrophage-like population, displaying molecular features distinct from those of tonsil CD11b+ CD14+ -macrophages and cDC2. These findings provide a novel view on the terminal differentiation path of slan+ -monocytes, which is relevant for inflammatory diseases and lymphomas.


Subject(s)
Amino Sugars/metabolism , Dendritic Cells/metabolism , Macrophages/metabolism , Monocytes/metabolism , Palatine Tonsil/metabolism , Tonsillitis/genetics , Case-Control Studies , Cells, Cultured , Dendritic Cells/cytology , Gene Expression Profiling , Humans , Macrophages/cytology , Monocytes/cytology , Palatine Tonsil/cytology , Tonsillitis/metabolism , Tonsillitis/pathology
6.
Eur J Immunol ; 49(10): 1457-1973, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31633216

ABSTRACT

These guidelines are a consensus work of a considerable number of members of the immunology and flow cytometry community. They provide the theory and key practical aspects of flow cytometry enabling immunologists to avoid the common errors that often undermine immunological data. Notably, there are comprehensive sections of all major immune cell types with helpful Tables detailing phenotypes in murine and human cells. The latest flow cytometry techniques and applications are also described, featuring examples of the data that can be generated and, importantly, how the data can be analysed. Furthermore, there are sections detailing tips, tricks and pitfalls to avoid, all written and peer-reviewed by leading experts in the field, making this an essential research companion.


Subject(s)
Allergy and Immunology/standards , Cell Separation/methods , Cell Separation/standards , Flow Cytometry/methods , Flow Cytometry/standards , Consensus , Humans , Phenotype
7.
Blood ; 129(10): 1343-1356, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28053192

ABSTRACT

The identification of discrete neutrophil populations, as well as the characterization of their immunoregulatory properties, is an emerging topic under extensive investigation. In such regard, the presence of circulating CD66b+ neutrophil populations, exerting either immunosuppressive or proinflammatory functions, has been described in several acute and chronic inflammatory conditions. However, due to the lack of specific markers, the precise phenotype and maturation status of these neutrophil populations remain unclear. Herein, we report that CD10, also known as common acute lymphoblastic leukemia antigen, neutral endopeptidase, or enkephalinase, can be used as a marker that, within heterogeneous populations of circulating CD66b+ neutrophils present in inflammatory conditions, clearly distinguishes the mature from the immature ones. Accordingly, we observed that the previously described immunosuppressive neutrophil population that appears in the circulation of granulocyte colony-stimulating factor (G-CSF)-treated donors (GDs) consists of mature CD66b+CD10+ neutrophils displaying an activated phenotype. These neutrophils inhibit proliferation and interferon γ (IFNγ) production by T cells via a CD18-mediated contact-dependent arginase 1 release. By contrast, we found that immature CD66b+CD10- neutrophils, also present in GDs, display an immature morphology, promote T-cell survival, and enhance proliferation and IFNγ production by T cells. Altogether, our findings uncover that in GDs, circulating mature and immature neutrophils, distinguished by their differential CD10 expression, exert opposite immunoregulatory properties. Therefore, CD10 might be used as a phenotypic marker discriminating mature neutrophils from immature neutrophil populations present in patients with acute or chronic inflammatory conditions, as well as facilitating their isolation, to better define their specific immunoregulatory properties.


Subject(s)
Biomarkers/analysis , Lymphocyte Activation/immunology , Neprilysin/biosynthesis , Neutrophils/immunology , T-Lymphocytes/immunology , Cell Separation , Flow Cytometry , Granulocyte Colony-Stimulating Factor/immunology , Humans , Neprilysin/analysis , Neprilysin/immunology
9.
Eur J Clin Invest ; 48 Suppl 2: e12952, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29772063

ABSTRACT

Polymorphonuclear neutrophils are the most numerous leucocytes present in human blood, and function as crucial players in innate immune responses. Neutrophils are indispensable for the defence towards microbes, as they effectively counter them by releasing toxic enzymes, by synthetizing reactive oxygen species and by producing inflammatory mediators. Interestingly, recent findings have highlighted an important role of neutrophils also as promoters of the resolution of inflammation process, indicating that their biological functions go well beyond simple pathogen killing. Consistently, data from the last decades have highlighted that neutrophils may even contribute to the development of adaptive immunity by performing previously unanticipated functions, including the capacity to extend their survival, directly interact with other leucocytes or cell types, and produce and release a variety of cytokines. In this article, we will summarize the main features of, as well as emphasize some important concepts on, the production of cytokines by human neutrophils.


Subject(s)
Cytokines/biosynthesis , Neutrophils/metabolism , Cell Survival , Humans , Interleukin-17/metabolism , Neutrophils/physiology , Protein Biosynthesis/physiology
10.
J Allergy Clin Immunol ; 136(3): 781-791.e9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26044852

ABSTRACT

BACKGROUND: IL-10 is well known for its ability to block the expression and production of numerous proinflammatory cytokines, in this manner preventing the development of excessive or chronic immune activation. IL-10-induced transcriptional repression of CXCL8 and TNFA genes consists of 2 distinct phases: an early phase, occurring rapidly and in a protein synthesis-independent manner, followed by a second phase that is more delayed and dependent on protein synthesis. OBJECTIVE: We sought to identify the mechanisms through which IL-10 rapidly and directly suppresses LPS-induced CXCL8 and TNF-α transcription, which might be defective under pathologic conditions. METHODS: The molecular events triggered by IL-10 in LPS-activated monocytes at the CXCL8 and TNFA loci were investigated by using the chromatin immunoprecipitation assay. RESULTS: Inhibition of LPS-induced CXCL8 and TNF-α expression by IL-10 proceeds through a common mechanism targeting LPS-induced phosphorylation of the nuclear factor κB p65 serine 276 residue (pS276p65). As a result, all the pS276p65-dependent events occurring at the CXCL8 and TNFA loci are consistently reduced, ultimately leading to a reduction in transcript elongation. Additionally, IL-10 selectively controls CXCL8 transcript elongation through histone deacetylase (HDAC) 2-dependent covalent chromatin modifications, disrupting the assembly of the transcriptional machinery. Remarkably, PBMCs from patients with acute-phase chronic obstructive pulmonary disease, which express negligible HDAC2 levels, are scarcely affected by IL-10 in terms of inhibition of CXCL8 expression. CONCLUSIONS: This study provides mechanistic evidence that IL-10 creates a chromatin environment that decreases the transcriptional rate of CXCL8 and TNF-α to Toll-like receptor 4-activating signals. Data identify novel molecular targets for therapeutic strategies aimed at dampening inflammation in pathologies such as chronic obstructive pulmonary disease, in which reduced intracellular HDAC2 levels have been described.


Subject(s)
Interleukin-10/immunology , Interleukin-8/immunology , Monocytes/immunology , Nuclear Proteins/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Transcription Factors/immunology , Tumor Necrosis Factor-alpha/immunology , Adult , Aged , Binding Sites , Case-Control Studies , Cell Cycle Proteins , Female , Gene Expression Regulation , Histone Deacetylase 2/genetics , Histone Deacetylase 2/immunology , Humans , Interleukin-10/metabolism , Interleukin-10/pharmacology , Interleukin-8/genetics , Lipopolysaccharides/pharmacology , Male , Middle Aged , Monocytes/drug effects , Monocytes/pathology , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Primary Cell Culture , Protein Binding , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/pathology , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription, Genetic , Tumor Necrosis Factor-alpha/genetics
11.
J Immunol ; 189(3): 1500-9, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22730532

ABSTRACT

Upon LPS binding, TLR4 activates a MyD88-dependent pathway leading to the transcriptional activation of proinflammatory genes, as well as a MyD88-independent/TRIF-dependent pathway, responsible for the transcriptional induction of IFN-ß. Previous findings delineated that human neutrophils are unable to induce the transcription of IFN-ß in response to TLR4 stimulation. Because neutrophils do not express protein kinase C ε, a molecule recently reported as essential for initiating the MyD88-independent/TRIF-dependent pathway, we optimized an electroporation method to transfect PKCε into neutrophils with very high efficiency. By doing so, a significant IFN-ß mRNA expression was induced, in the absence of LPS stimulation, not only in PKCε-overexpressing neutrophils but also in cells transfected with a series of empty DNA plasmids; however, LPS further upregulated the IFN-ß transcript levels in plasmid-transfected neutrophils, regardless of PKCε overexpression. Phosphoimmunoblotting studies, as well as chromatin immunoprecipitation assays targeting the IFN-ß promoter, revealed that IFN-ß mRNA induction occurred through the cooperative action of IRF3, activated by transfected DNA, and NF-κB, activated by LPS. Additional immunoblotting and coimmunoprecipitation studies revealed that neutrophils constitutively express various cytosolic DNA sensors, including IFN-inducible protein 16, leucine-rich repeat (in Flightless I) interacting protein-1, and DDX41, as well as that IFN-inducible protein 16 is the intracellular receptor recognizing transfected DNA. Consistently, infection of neutrophils with intracellular pathogens, such as Bartonella henselae, Listeria monocytogenes, Legionella pneumophila, or adenovirus type 5, promoted a marked induction of IFN-ß mRNA expression. Taken together, these data raise questions about the role of PKCε in driving the MyD88-independent/TRIF-dependent response and indicate that human neutrophils are able to recognize and respond to microbial cytosolic DNA.


Subject(s)
DNA/biosynthesis , Interferon-beta/biosynthesis , Neutrophils/immunology , Plasmids/genetics , Signal Transduction/immunology , Toll-Like Receptor 4/physiology , Transcriptional Activation/immunology , Up-Regulation/immunology , Adenoviruses, Human/genetics , Adenoviruses, Human/immunology , Bartonella henselae/genetics , Bartonella henselae/immunology , Cells, Cultured , Cytosol/immunology , DNA/genetics , HEK293 Cells , Humans , Interferon-beta/genetics , Legionella pneumophila/genetics , Legionella pneumophila/immunology , Listeria monocytogenes/genetics , Listeria monocytogenes/immunology , Neutrophils/metabolism , Neutrophils/microbiology , RNA, Messenger/biosynthesis , Signal Transduction/genetics , Transfection/methods , Up-Regulation/genetics
13.
J Leukoc Biol ; 115(4): 695-705, 2024 03 29.
Article in English | MEDLINE | ID: mdl-38114064

ABSTRACT

The advent of recent cutting-edge technologies has allowed the discovery and characterization of novel progenitors of human neutrophils, including SSCloCD66b+CD15+CD11b-CD49dhiproNeu1s, SSChiCD66b+CD15+CD11b-CD49dintproNeus2s, CD66b+CD15+CD11b+CD49d+CD101-preNeus, and Lin-CD66b+CD117+CD71+eNePs. In this research field, we recently identified CD66b-CD38+CD64dimCD115-, CD34+, and CD34dim/- cells exclusively committed to the neutrophil lineage (which we renamed as CD34+ and CD34dim/- neutrophil-committed progenitors), representing the earliest neutrophil precursors identifiable and sorted by flow cytometry. Moreover, based on their differential CD34 and CD45RA expression, we could identify 4 populations of neutrophil-committed progenitors: CD34+CD45RA-/NCP1s, CD34+CD45RA+/NCP2s, CD34dim/-CD45RA+/NCP3s, and CD34dim/-CD45RA-/NCP4s. This said, a very recent study by Ikeda and coworkers (PMID: 36862552) reported that neutrophil precursors, termed either neutrophil progenitors or "early neutrophil-committed progenitors," would generate immunosuppressive neutrophil-like CXCR1+CD14+CD16- monocytes. Hence, presuming that neutrophil progenitors/"early neutrophil-committed progenitors" correspond to neutrophil-committed progenitors, the selective neutrophil commitment that we attributed to neutrophil-committed progenitors is contradicted by Ikeda and coworkers' article. In this study, by performing a more analytical reevaluation at the phenotypic and molecular levels of the cells generated by neutrophil-committed progenitors 2 and 4 (selected as representatives of neutrophil-committed progenitors), we categorically exclude that neutrophil-committed progenitors generate neutrophil-like CXCR1+CD14+CD16- monocytes. Rather, we provide substantial evidence indicating that the cells generated by neutrophil progenitors/"early neutrophil-committed progenitors" are neutrophilic cells at a different stage of maturation, displaying moderate levels of CD14, instead of neutrophil-like CXCR1+CD14+CD16- monocytes, as pointed by Ikeda and coworkers. Hence, the conclusion that neutrophil progenitors/"early neutrophil-committed progenitors" aberrantly differentiate into neutrophil-like monocytes derives, in our opinion, from data misinterpretation.


Subject(s)
Monocytes , Neutrophils , Humans , Neutrophils/metabolism , Monocytes/metabolism , Antigens, CD34/metabolism , Flow Cytometry
14.
Cell Rep Med ; 5(2): 101380, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38242120

ABSTRACT

Precise molecular characterization of circulating polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) is hampered by their mixed composition of mature and immature cells and lack of specific markers. Here, we focus on mature CD66b+CD10+CD16+CD11b+ PMN-MDSCs (mPMN-MDSCs) from either cancer patients or healthy donors receiving G-CSF for stem cell mobilization (GDs). By RNA sequencing (RNA-seq) experiments, we report the identification of a distinct gene signature shared by the different mPMN-MDSC populations under investigation, also validated in mPMN-MDSCs from GDs and tumor-associated neutrophils (TANs) by single-cell RNA-seq (scRNA-seq) experiments. Analysis of such a gene signature uncovers a specific transcriptional program associated with mPMN-MDSC differentiation and allows us to identify that, in patients with either solid or hematologic tumors and in GDs, CD52, CD84, and prostaglandin E receptor 2 (PTGER2) represent potential mPMN-MDSC-associated markers. Altogether, our findings indicate that mature PMN-MDSCs distinctively undergo specific reprogramming during differentiation and lay the groundwork for selective immunomonitoring, and eventually targeting, of mature PMN-MDSCs.


Subject(s)
Myeloid-Derived Suppressor Cells , Neoplasms , Humans , Neutrophils , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte Colony-Stimulating Factor/metabolism , Neoplasms/pathology , CD52 Antigen/metabolism , Signaling Lymphocytic Activation Molecule Family/metabolism
15.
Blood ; 117(5): 1677-86, 2011 Feb 03.
Article in English | MEDLINE | ID: mdl-21098395

ABSTRACT

The role of neutrophils as key players in the regulation of innate and adaptive immune responses is increasingly being recognized. We report that human neutrophils establish a network with both natural killer (NK) cells and 6-sulfo LacNAc(+) dendritic cells (slanDCs), which ultimately serves to up-regulate NK-derived interferonγ (IFNγ). This network involves direct reciprocal interactions and positive amplification loops mediated by cell-derived cytokines. Accordingly, we show that after lipopolysaccharide + interleukin-2 (IL-2) or IL-15/IL-18 stimulation, neutrophils directly interact with and potentiate the activity of both slanDCs and NK cells. On the one hand, neutrophils augment the release of IL-12p70 by slanDCs via a CD18/ intercellular adhesion molecule-1 (ICAM-1) interaction that stimulates activated NK cells to produce IFNγ. IFNγ further potentiates the interaction between neutrophils and slanDCs and the release of slanDC-derived IL-12p70, thus creating a positive feedback loop. On the other hand, neutrophils directly co-stimulate NK cells via CD18/ICAM-3, leading to the production of IFNγ. Colocalization of neutrophils, NK cells, and slanDCs, as well as of IL-12p70 and IFNγ, in inflamed tissues of Crohn disease and psoriasis provides strong evidence for a novel cellular and cytokine cooperation within the innate immune system in which neutrophils act as amplifiers of NK cell/slanDC-mediated responses.


Subject(s)
Amino Sugars/metabolism , Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Dendritic Cells/metabolism , Intercellular Adhesion Molecule-1/metabolism , Interferon-gamma/metabolism , Interleukin-18/metabolism , Killer Cells, Natural/metabolism , Neutrophils/metabolism , Cells, Cultured , Crohn Disease/immunology , Crohn Disease/metabolism , Crohn Disease/pathology , Dendritic Cells/cytology , Humans , Inflammation/metabolism , Inflammation/pathology , Interleukin-15/metabolism , Interleukin-2/metabolism , Killer Cells, Natural/cytology , Neutrophils/cytology , Psoriasis/immunology , Psoriasis/metabolism , Psoriasis/pathology
17.
Front Immunol ; 14: 1287656, 2023.
Article in English | MEDLINE | ID: mdl-37965335

ABSTRACT

Introduction: Peripheral monocytes in humans are conventionally divided into classical (CL, CD14++CD16-), intermediate (INT, CD14++CD16+) and non-classical (NC, CD14dim/-CD16++) cells, based on their expression levels of CD14 and CD16. A major fraction of the NC-monocytes has been shown to express the 6-sulfo LacNAc (slan) antigen, but whether these slan+/NC-monocytes represent the prototypical non-classical monocytes or whether they are simply a sub-fraction with identical features as the remainder of NC monocytes is still unclear. Methods: We analyzed transcriptome (by bulk and single cell RNA-seq), proteome, cell surface markers and production of discrete cytokines by peripheral slan+/NC- and slan-/NC-monocytes, in comparison to total NC-, CL- and INT- monocytes. Results: By bulk RNA-seq and proteomic analysis, we found that slan+/NC-monocytes express higher levels of genes and proteins specific of NC-monocytes than slan-/NC-monocytes do. Unsupervised clustering of scRNA-seq data generated one cluster of NC- and one of INT-monocytes, where all slan+/NC-monocytes were allocated to the NC-monocyte cluster, while slan-/NC-monocytes were found, in part (13.4%), within the INT-monocyte cluster. In addition, total NC- and slan-/NC-monocytes, but not slan+/NC-monocytes, were found by both bulk RNA-seq and scRNA-seq to contain a small percentage of natural killer cells. Conclusion: In addition to comparatively characterize total NC-, slan-/NC- and slan+/NC-monocyte transcriptomes and proteomes, our data prove that slan+/NC-, but not slan-/NC-, monocytes are more representative of prototypical NC-monocytes.


Subject(s)
Monocytes , Proteomics , Humans , Leukocytes, Mononuclear
18.
Stem Cells ; 29(6): 1001-11, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21563279

ABSTRACT

Bone marrow-derived mesenchymal stromal cells (BM-MSCs) are stromal precursors endowed with extensive immunomodulative properties. In this study, we aimed to assess whether Toll-like receptor-3 (TLR3)- and TLR4-activated BM-MSC influence human neutrophil (PMN) responses under coculture conditions. We show that TLR3 triggering by polyinosinic:polycytidylic acid dramatically amplifies, in a more significant manner than TLR4 triggering by lipopolysaccharide, the antiapoptotic effects that resting BM-MSC constitutively exert on PMN under coculture conditions, preserving a significant fraction of viable and functional PMN up to 72 hours. In addition, TLR3- and TLR4-activated BM-MSC enhance respiratory burst ability and CD11b expression by PMN. The coculture in the absence of cell contact and the incubation of PMN in supernatants harvested from TLR3- and TLR4-activated BM-MSC yield comparable results in terms of increased survival and immunophenotypic changes, thus suggesting the involvement of endogenous soluble factors. Neutralizing experiments reveal that the biological effects exerted on PMN by TLR3-activated BM-MSC are mediated by the combined action of interleukin 6, interferon-ß (IFN-ß), and granulocyte macrophage colony-stimulating factor (GM-CSF), while those exerted by TLR4-activated BM-MSC mostly depend on GM-CSF. MSC isolated from thymus, spleen, and subcutaneous adipose tissue behaves similarly. Finally, the effects exerted by TLR3- or TLR4-stimulated BM-MSC on PMN are conserved even after the previous priming of BM-MSC with IFN-γ and tumor necrosis factor-α. Our data highlight a novel mechanism by which MSC sustain and amplify the functions of PMN in response to TLR3- and TLR4-triggering and may consequently contribute to inflammatory disorders.


Subject(s)
Cell Survival/drug effects , Mesenchymal Stem Cells/metabolism , Neutrophils/physiology , Toll-Like Receptor 3/metabolism , Bone Marrow Cells/cytology , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned , Granulocyte Colony-Stimulating Factor/metabolism , Humans , Interferon-beta/metabolism , Interferon-gamma/pharmacology , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Mesenchymal Stem Cells/drug effects , Neutrophils/drug effects , Phenotype , Poly I-C/pharmacology , Toll-Like Receptor 3/agonists , Tumor Necrosis Factor-alpha/pharmacology
19.
Eur J Immunol ; 40(3): 722-32, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20039296

ABSTRACT

The CD300e surface molecule, originally termed immune receptor expressed by myeloid cells (IREM)-2, was reported to associate with the DNAX-activating protein (DAP) 12 adaptor in co-transfected cells, and is capable of signaling. In the present report, we investigated in detail the function of CD300e in monocytes and myeloid DC (mDC) freshly isolated from peripheral blood of normal blood donors. Upon engagement by an agonistic mAb, CD300e triggered an intracellular calcium mobilization and superoxide anion O(2) (-) production in monocytes. Activation via CD300e provided survival signals that prevented monocyte and mDC apoptosis, triggered the production of pro-inflammatory cytokines and upregulated the expression of cell surface co-stimulatory molecules in both cell types. Moreover, CD300e activation of mDC enhanced the alloreactive response of naive T cells. Overall, our data formally support the notion that CD300e functions as an activating receptor capable of regulating the innate immune response in myeloid cells.


Subject(s)
Dendritic Cells/immunology , Monocytes/immunology , Myeloid Cells/immunology , Receptors, Immunologic/immunology , Signal Transduction/immunology , Antigen Presentation/immunology , Cell Line , Cell Separation , Dendritic Cells/metabolism , Flow Cytometry , Humans , Lymphocyte Activation/immunology , Monocytes/metabolism , Myeloid Cells/metabolism , Receptors, Immunologic/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
20.
Haematologica ; 96(10): 1543-7, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21712539

ABSTRACT

Interaction between neutrophils and other leukocytes plays a variety of important roles in regulating innate and adaptive immune responses. Recently, we have shown that neu-trophils amplify NK cell/6-sulfo LacNAc(+) dendritic cells (slanDC)-mediated cytokine production, by potentiating IL-12p70 release by slanDC via CD18/ICAM-1 and directly co-stimulating IFNγ production by NK cells via ICAM-3. Herein, we have identified additional molecules involved in the interactions among neutrophils, NK cells and slanDC. More specifically, we provide evidence that: i) the cross-talk between neutrophils and NK cells is mediated by ICAM-3 and CD11d/CD18, respectively; ii) slanDC potentiate the production of IFNγ by NK cells via CD11a/CD18. Altogether, our studies shed more light on the role that adhesion molecules play within the neutrophil/NK cell/slanDC network. Our data also have potential implications in the pathogenesis of diseases driven by hyperactivated leukocytes, such as Sweet's syndrome, in which a neutrophil/NK cell co-localization is frequently observed.


Subject(s)
Antigens, CD/metabolism , CD11 Antigens/metabolism , Cell Adhesion Molecules/metabolism , Integrin alpha Chains/metabolism , Interferon-gamma/biosynthesis , Killer Cells, Natural/immunology , Neutrophils/immunology , CD11a Antigen/metabolism , CD18 Antigens/metabolism , Cell Communication , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Interleukin-12/biosynthesis , Killer Cells, Natural/metabolism , Lectins, C-Type/metabolism , Neutrophils/metabolism , Protein Binding , Protein Transport , Receptors, Cell Surface/metabolism , Sweet Syndrome/immunology , Sweet Syndrome/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL