Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
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.
Nat Immunol ; 23(10): 1470-1483, 2022 10.
Article in English | MEDLINE | ID: mdl-36138183

ABSTRACT

Traditionally viewed as poorly plastic, neutrophils are now recognized as functionally diverse; however, the extent and determinants of neutrophil heterogeneity in humans remain unclear. We performed a comprehensive immunophenotypic and transcriptome analysis, at a bulk and single-cell level, of neutrophils from healthy donors and patients undergoing stress myelopoiesis upon exposure to growth factors, transplantation of hematopoietic stem cells (HSC-T), development of pancreatic cancer and viral infection. We uncover an extreme diversity of human neutrophils in vivo, reflecting the rates of cell mobilization, differentiation and exposure to environmental signals. Integrated control of developmental and inducible transcriptional programs linked flexible granulopoietic outputs with elicitation of stimulus-specific functional responses. In this context, we detected an acute interferon (IFN) response in the blood of patients receiving HSC-T that was mirrored by marked upregulation of IFN-stimulated genes in neutrophils but not in monocytes. Systematic characterization of human neutrophil plasticity may uncover clinically relevant biomarkers and support the development of diagnostic and therapeutic tools.


Subject(s)
Myelopoiesis , Neutrophils , Biomarkers/metabolism , Humans , Interferons/genetics , Interferons/metabolism , Neutrophils/metabolism , Plastics/metabolism
3.
Trends Immunol ; 41(12): 1059-1061, 2020 12.
Article in English | MEDLINE | ID: mdl-33160842

ABSTRACT

Recent studies have revealed that neutrophils exhibit an unsuspected heterogeneity. In this context, the term high-density neutrophils (HDNs) has recently gained ground to define nothing more than neutrophils displaying an unaltered normal density. Therefore, as discussed here, we argue that the HDNs term must be avoided, as it is confounding and scientifically inappropriate.


Subject(s)
Neutrophils , Terminology as Topic , Cell Count , Humans , Neutrophils/cytology
4.
Trends Immunol ; 40(7): 565-583, 2019 07.
Article in English | MEDLINE | ID: mdl-31160207

ABSTRACT

New evidence has challenged the outdated dogma that neutrophils are a homogeneous population of short-lived cells. Although neutrophil subpopulations with distinct functions have been reported under homeostatic and pathological conditions, a full understanding of neutrophil heterogeneity and plasticity is currently lacking. We review here current knowledge of neutrophil heterogeneity and diversity, highlighting the need for deep genomic, phenotypic, and functional profiling of the identified neutrophil subpopulations to determine whether these cells truly represent bona fide novel neutrophil subsets. We suggest that progress in understanding neutrophil heterogeneity will allow the identification of clinically relevant neutrophil subpopulations that may be used in the diagnosis of specific diseases and lead to the development of new therapeutic approaches.


Subject(s)
Cell Plasticity , Disease Susceptibility , Homeostasis , Neutrophils/immunology , Neutrophils/metabolism , Phenotype , Animals , Biomarkers , Female , Humans , Immunity, Innate , Immunomodulation , Leukocyte Count , Neutrophils/pathology , Pregnancy
5.
Eur J Immunol ; 49(8): 1213-1225, 2019 08.
Article in English | MEDLINE | ID: mdl-31034584

ABSTRACT

Among the family of regulatory B cells, the subset able to produce interleukin-10 (IL-10) is the most studied, yet its biology is still a matter of investigation. The DNA methylation profiling of the il-10 gene locus revealed a novel epigenetic signature characterizing murine B cells ready to respond through IL-10 synthesis: a demethylated region located 4.5 kb from the transcription starting site (TSS), that we named early IL10 regulatory region (eIL10rr). This feature allows to distinguish B cells that are immediately prone and developmentally committed to IL-10 production from those that require a persistent stimulation to exert an IL-10-mediated regulatory function. These late IL-10 producers are instead characterized by a delayed IL10 regulatory region (dIL10rr), a partially demethylated DNA portion located 9 kb upstream from the TSS. A demethylated region was also found in human IL-10-producing B cells and, very interestingly, in some B-cell malignancies, such as chronic lymphocytic leukemia and mantle cell lymphoma, characterized by an immunosuppressive microenvironment. Our findings define murine and human regulatory B cells as an epigenetically controlled functional state of mature B cell subsets and open a new perspective on IL-10 regulation in B cells in homeostasis and disease.


Subject(s)
B-Lymphocyte Subsets/physiology , B-Lymphocytes, Regulatory/physiology , Interleukin-10/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Lymphoma, Mantle-Cell/genetics , Regulatory Sequences, Nucleic Acid/genetics , Animals , Cell Differentiation , DNA Methylation , Female , Gene Expression Profiling , Humans , Immune Tolerance , Immunity, Humoral , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Tumor Microenvironment
6.
Immunol Rev ; 273(1): 48-60, 2016 09.
Article in English | MEDLINE | ID: mdl-27558327

ABSTRACT

Recent findings have uncovered novel fascinating aspects of the biology of neutrophils, which ultimately attribute to these cells a broader role in inflammation and immunity. One aspect that is currently under intensive investigation is the notion of neutrophil 'heterogeneity'. Studies examining neutrophils in a variety of acute and chronic inflammatory conditions report, in fact, the recovery of CD66b(+) cells displaying neutrophil-like morphology at different degrees of maturation/activation, able to exert either immunosuppressive or proinflammatory properties. These heterogeneous populations of mature and immature neutrophils are indicated with a variety of names, including 'low density neutrophils (LDNs)', 'low density granulocytes (LDGs)', 'granulocytic-myeloid derived suppressor cells (G-MDSCs)', and immunosuppressive neutrophils. However, due to the lack of discrete markers that can unequivocally allow their specific identification and isolation, the precise phenotype and function of all these presumably novel, neutrophil-like, populations have not been correctly defined yet. Aim of this article is to summarize current knowledge on the mature and immature neutrophil populations described to date, featuring immunosuppressive or proinflammatory properties, often defined as 'subsets', as well as to critically discuss unresolved issues in the field.


Subject(s)
Cell Differentiation , Immunity, Innate , Immunosuppression Therapy , Inflammation/immunology , Neutrophils/physiology , Animals , Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Cell Lineage , GPI-Linked Proteins/metabolism , Humans , Neutrophil Activation
7.
Immunology ; 156(1): 23-32, 2019 01.
Article in English | MEDLINE | ID: mdl-30259972

ABSTRACT

An increasing body of literature supports a role for neutrophils as players in the orchestration of adaptive immunity. During acute and chronic inflammatory conditions, neutrophils rapidly migrate not only to sites of inflammation, but also to draining lymph nodes and spleen, where they engage bidirectional interactions with B- and T-lymphocyte subsets. Accordingly, a relevant role of neutrophils in modulating B-cell responses under homeostatic conditions has recently emerged. Moreover, specialized immunoregulatory properties towards B or T cells acquired by distinct neutrophil populations, originating under pathological conditions, have been consistently described. In this article, we summarize the most recent data from human studies and murine models on the ability of neutrophils to modulate adaptive immune responses under physiological and pathological conditions and the mechanisms behind these processes.


Subject(s)
B-Lymphocytes/immunology , Neutrophils/immunology , T-Lymphocytes/immunology , Adaptive Immunity , Animals , Cell Communication , Homeostasis , Humans , Immunity, Innate , Mice
8.
Cancer Immunol Immunother ; 68(4): 687-697, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30684003

ABSTRACT

In cancer, infection and inflammation, the immune system's function can be dysregulated. Instead of fighting disease, immune cells may increase pathology and suppress host-protective immune responses. Myeloid cells show high plasticity and adapt to changing conditions and pathological challenges. Despite their relevance in disease pathophysiology, the identity, heterogeneity and biology of myeloid cells is still poorly understood. We will focus on phenotypical and functional markers of one of the key myeloid regulatory subtypes, the myeloid derived suppressor cells (MDSC), in humans, mice and non-human primates. Technical issues regarding the isolation of the cells from tissues and blood, timing and sample handling of MDSC will be detailed. Localization of MDSC in a tissue context is of crucial importance and immunohistochemistry approaches for this purpose are discussed. A minimal antibody panel for MDSC research is provided as part of the Mye-EUNITER COST action. Strategies for the identification of additional markers applying state of the art technologies such as mass cytometry will be highlighted. Such marker sets can be used to study MDSC phenotypes across tissues, diseases as well as species and will be crucial to accelerate MDSC research in health and disease.


Subject(s)
Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Animals , Biomarkers , Cell Separation/methods , Humans , Immunophenotyping/methods , Mice , Neutrophils/immunology , Neutrophils/metabolism , Primates
9.
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
10.
Immunity ; 33(2): 181-91, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20705491

ABSTRACT

A20 is a ubiquitin modifying enzyme that restricts NF-kappaB signals and protects cells against tumor necrosis factor (TNF)-induced programmed cell death. Given recent data linking A20 (TNFAIP3) with human B cell lymphomas and systemic lupus erythematosus (SLE), we have generated mice bearing a floxed allele of Tnfaip3 to interrogate A20's roles in regulating B cell functions. A20-deficient B cells are hyperresponsive to multiple stimuli and display exaggerated NF-kappaB responses to CD40-induced signals. Mice expressing absent or hypomorphic amounts of A20 in B cells possess elevated numbers of germinal center B cells, autoantibodies, and glomerular immunoglobulin deposits. A20-deficient B cells are resistant to Fas-mediated cell death, probably due to increased expression of NF-kappaB-dependent antiapoptotic proteins such as Bcl-x. These findings show that A20 can restrict B cell survival, whereas A20 protects other cells from TNF-induced cell death. Our studies demonstrate how reduced A20 expression predisposes to autoimmunity.


Subject(s)
Autoimmunity , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cysteine Endopeptidases/immunology , Cysteine Endopeptidases/metabolism , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Animals , B-Lymphocytes/enzymology , CD40 Antigens/immunology , CD40 Antigens/metabolism , Cell Lineage , Cell Survival , Cysteine Endopeptidases/deficiency , Homeostasis , Intracellular Signaling Peptides and Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Signal Transduction , Tumor Necrosis Factor alpha-Induced Protein 3
11.
Blood ; 124(5): 710-9, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-24923297

ABSTRACT

It is now widely recognized that neutrophils are highly versatile and sophisticated cells that display de novo synthetic capacity and may greatly extend their lifespan. In addition, concepts such as "neutrophil heterogeneity" and "neutrophil plasticity" have started to emerge, implying that, under pathological conditions, neutrophils may differentiate into discrete subsets defined by distinct phenotypic and functional profiles. A number of studies have shown that neutrophils act as effectors in both innate and adaptive immunoregulatory networks. In fact, once recruited into inflamed tissues, neutrophils engage into complex bidirectional interactions with macrophages, natural killer, dendritic and mesenchymal stem cells, B and T lymphocytes, or platelets. As a result of this cross-talk, mediated either by contact-dependent mechanisms or cell-derived soluble factors, neutrophils and target cells reciprocally modulate their survival and activation status. Altogether, these novel aspects of neutrophil biology have shed new light not only on the potential complex roles that neutrophils play during inflammation and immune responses, but also in the pathogenesis of several inflammatory disorders including infection, autoimmunity, and cancer.


Subject(s)
Adaptive Immunity/physiology , Cell Communication/physiology , Immunity, Innate/physiology , Neutrophils/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Humans , Infections/immunology , Infections/pathology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Macrophages/cytology , Macrophages/immunology , Neoplasms/immunology , Neoplasms/pathology , Neutrophils/cytology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
12.
J Immunol ; 192(3): 875-85, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24379120

ABSTRACT

The intracellular tyrosine kinase Lyn mediates inhibitory receptor function in B cells and myeloid cells, and Lyn(-/-) mice spontaneously develop an autoimmune and inflammatory disease that closely resembles human systemic lupus erythematosus. TLR-signaling pathways have been implicated in the production of anti-nuclear Abs in systemic lupus erythematosus and mouse models of it. We used a conditional allele of Myd88 to determine whether the autoimmunity of Lyn(-/-) mice is dependent on TLR/MyD88 signaling in B cells and/or in dendritic cells (DCs). The production of IgG anti-nuclear Abs, as well as the deposition of these Abs in the glomeruli of the kidneys, leading to glomerulonephritis in Lyn(-/-) mice, were completely abolished by selective deletion of Myd88 in B cells, and autoantibody production and glomerulonephritis were delayed or decreased by deletion of Myd88 in DCs. The reduced autoantibody production in mice lacking MyD88 in B cells or DCs was accompanied by a dramatic decrease in the spontaneous germinal center (GC) response, suggesting that autoantibodies in Lyn(-/-) mice may depend on GC responses. Consistent with this view, IgG anti-nuclear Abs were absent if T cells were deleted (TCRß(-/-) TCRδ(-/-) mice) or if T cells were unable to contribute to GC responses as the result of mutation of the adaptor molecule SAP. Thus, the autoimmunity of Lyn(-/-) mice was dependent on T cells and on TLR/MyD88 signaling in B cells and in DCs, supporting a model in which DC hyperactivity combines with defects in tolerance in B cells to lead to a T cell-dependent systemic autoimmunity in Lyn(-/-) mice.


Subject(s)
Antibodies, Antinuclear/biosynthesis , B-Lymphocytes/immunology , Dendritic Cells/immunology , Germinal Center/immunology , Immunoglobulin G/biosynthesis , Lupus Nephritis/immunology , Myeloid Differentiation Factor 88/physiology , src-Family Kinases/deficiency , Animals , Antibodies, Antinuclear/genetics , Antibodies, Antinuclear/immunology , Antigen-Antibody Complex/analysis , Disease Models, Animal , Gene Deletion , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Intracellular Signaling Peptides and Proteins/physiology , Lupus Erythematosus, Systemic , Lupus Nephritis/pathology , Lymphocyte Count , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Receptors, Antigen, T-Cell, gamma-delta/deficiency , Self Tolerance/immunology , Signal Transduction/immunology , Signaling Lymphocytic Activation Molecule Associated Protein , Specific Pathogen-Free Organisms , Toll-Like Receptors/immunology
13.
Proc Natl Acad Sci U S A ; 110(35): E3311-20, 2013 Aug 27.
Article in English | MEDLINE | ID: mdl-23940344

ABSTRACT

Deletion of lyn, a Src-family tyrosine kinase expressed by B, myeloid, and dendritic cells (DCs), triggers lupus-like disease in mice, characterized by autoantibody production and renal immune complex deposition leading to chronic glomerulonephritis. B cells from these mice are hyperactive to antigen-receptor stimulation owing to a loss of inhibitory signaling mediated by Lyn kinase. The hyperactive B-cell responses are thought to underlie the development of autoimmunity in this model. Lyn-deficient mice also manifest significant myeloexpansion. To test the contribution of different immune cell types to the lupus-like disease in this model, we generated a lyn(flox/flox) transgenic mouse strain. To our surprise, when we crossed these mice to Cd11c-cre animals, generating DC-specific deletion of Lyn, the animals developed spontaneous B- and T-cell activation and subsequent production of autoantibodies and severe nephritis. Remarkably, the DC-specific Lyn-deficient mice also developed severe tissue inflammatory disease, which was not present in the global lyn(-/-) strain. Lyn-deficient DCs were hyperactivated and hyperresponsive to Toll-like receptor agonists and IL-1ß. To test whether dysregulation of these signaling pathways in DCs contributed to the inflammatory/autoimmune phenotype, we crossed the lyn(f/f) Cd11c-cre(+) mice to myd88(f/f) animals, generating double-mutant mice lacking both Lyn and the adaptor protein myeloid differentiation factor 88 (MyD88) in DCs, specifically. Deletion of MyD88 in DCs alone completely reversed the inflammatory autoimmunity in the DC-specific Lyn-mutant mice. Thus, we demonstrate that hyperactivation of MyD88-dependent signaling in DCs is sufficient to drive pathogenesis of lupus-like disease, illuminating the fact that dysregulation in innate immune cells alone can lead to autoimmunity.


Subject(s)
Autoimmunity , Dendritic Cells/metabolism , Inflammation/metabolism , Myeloid Differentiation Factor 88/metabolism , Signal Transduction , src-Family Kinases/genetics , Animals , Autoantibodies/biosynthesis , Dendritic Cells/enzymology , Dendritic Cells/immunology , Lymphatic Diseases/genetics , Lymphatic Diseases/metabolism , Lymphocyte Activation , Mice , Mice, Knockout , Splenomegaly/genetics , Splenomegaly/metabolism
14.
Semin Cancer Biol ; 23(3): 159-70, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23410636

ABSTRACT

Although traditionally viewed as short-lived innate immunity cells, only playing a crucial role in host defense toward infections, neutrophils have recently become subject of a new wave of research in diverse areas including in tumors. Indeed, increasing experimental evidence indicate that neutrophils may directly or indirectly influence the tumor fate through the release of a wide array of molecules able to exert either pro-tumor or anti-tumor functions depending on the microenvironment milieu, including cytokines. This review therefore attempts to uncover the role that neutrophils play during the different steps of tumor development (from promotion to progression), as well as in anti-tumor responses, via cytokine production.


Subject(s)
Cytokines/biosynthesis , Neoplasms/immunology , Neoplasms/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Animals , Cytokines/immunology , Disease Progression , Humans , Neoplasms/pathology , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Neutrophil Infiltration/immunology
15.
Proc Natl Acad Sci U S A ; 108(41): E823-32, 2011 Oct 11.
Article in English | MEDLINE | ID: mdl-21911371

ABSTRACT

Lyn kinase deficient mice represent a well established genetic model of autoimmune/autoinflammatory disease that resembles systemic lupus erythematosus. We report that IL-10 plays a crucial immunosuppressive role in this model, modulating the inflammatory component of the disease caused by myeloid and T-cell activation. Double-mutant lyn(-/-)IL-10(-/-) mice manifested severe splenomegaly and lymphadenopathy, dramatically increased proinflammatory cytokine production, and severe tissue inflammation. Single-mutant lyn(-/-)mice showed expansion of IL-10-producing B cells. Interestingly, WT B cells adoptively transferred into lyn(-/-) mice showed increased differentiation into IL-10-producing B cells that assumed a similar phenotype to endogenous lyn(-/-) IL-10-producing B cells, suggesting that the inflammatory environment present in lyn(-/-) mice induces IL-10-producing B-cell differentiation. B cells, but not T or myeloid cells, were the critical source of IL-10 able to reduce inflammation and autoimmunity in double mutant lyn(-/-)IL-10(-/-) mice. IL-10 secretion by B cells was also crucial to sustain transcription factor Forkhead Box P3 (Foxp3) expression in regulatory T cells during disease development. These data reveal a dominant immunosuppressive function of B-cell-derived IL-10 in the Lyn-deficient model of autoimmunity, extending our current understanding of the role of IL-10 and IL-10-producing B cells in systemic lupus erythematosus.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Interleukin-10/physiology , Lupus Erythematosus, Systemic/prevention & control , src-Family Kinases/deficiency , Animals , B-Lymphocytes, Regulatory/enzymology , B-Lymphocytes, Regulatory/pathology , Cell Differentiation , Cytokines/biosynthesis , Disease Models, Animal , Humans , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Inflammation Mediators/metabolism , Interleukin-10/deficiency , Interleukin-10/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Lymphatic Diseases/etiology , Lymphatic Diseases/pathology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cells/immunology , Splenomegaly/etiology , Splenomegaly/pathology , T-Lymphocytes/immunology , src-Family Kinases/genetics
16.
J Leukoc Biol ; 115(4): 585-588, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38394343

ABSTRACT

Discoveries made in the past decades have brought out that, in addition to their classical primary defensive functions against infections, polymorphonuclear neutrophils play key effector roles not only in chronic inflammatory and immune-mediated diseases but also in cancer. In addition, depending on their differentiation/activation status and/or on the physiological or pathological microenvironment in which they reside, neutrophils have been shown to behave as highly plastic cells, able to acquire new phenotypes/functional states. All these features are well manifested in cancer and modulated during tumor progression. Herein, we discuss intriguing data by Lai Ng's group that have shed light on the origin and development of terminally differentiated, proangiogenic, tumor-associated neutrophils, facilitating tumor growth in a murine orthotopic model of pancreatic ductal adenocarcinoma. These findings help to progress toward the ambitious goal of selectively targeting only the skewed pathological neutrophil populations present within the tumor microenvironment.


Subject(s)
Neutrophils , Pancreatic Neoplasms , Humans , Animals , Mice , Neutrophils/pathology , Tumor Microenvironment/physiology
17.
J Leukoc Biol ; 115(2): 205-221, 2024 01 19.
Article in English | MEDLINE | ID: mdl-37824822

ABSTRACT

Cytosolic proliferating cell nuclear antigen (PCNA) is involved in neutrophil survival and function, in which it acts as a scaffold and associates with proteins involved in apoptosis, NADPH oxidase activation, cytoskeletal dynamics, and metabolism. While the PCNA interactome has been characterized in neutrophils under homeostatic conditions, less is known about neutrophil PCNA in pathophysiological contexts. Granulocyte colony-stimulating factor (G-CSF) is a cytokine produced in response to inflammatory stimuli that regulates many aspects of neutrophil biology. Here, we used isolated normal-density neutrophils from G-CSF-treated haemopoietic stem cell donors (GDs) as a model to understand the role of PCNA during inflammation. Proteomic analysis of the neutrophil cytosol revealed significant differences between GDs and healthy donors (HDs). PCNA was one of the most upregulated proteins in GDs, and the PCNA interactome was significantly different in GDs compared with HDs. Importantly, while PCNA associated with almost all enzymes involved in glycolysis in HDs, these associations were decreased in GDs. Functionally, neutrophils from GDs had a significant increase in glycolysis compared with HDs. Using p21 competitor peptides, we showed that PCNA negatively regulates neutrophil glycolysis in HDs but had no effect on GD neutrophils. These data demonstrate that G-CSF alters the PCNA scaffold, affecting interactions with key glycolytic enzymes, and thus regulates glycolysis, the main energy pathway utilized by neutrophils. By this selective control of glycolysis, PCNA can organize neutrophils functionality in parallel with other PCNA mechanisms of prolonged survival. PCNA may therefore be instrumental in the reprogramming that neutrophils undergo in inflammatory or tumoral settings.


Subject(s)
Granulocyte Colony-Stimulating Factor , Neutrophils , Neutrophils/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Cytosol/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Proteomics , Cytokines/metabolism
18.
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
19.
J Immunol ; 187(8): 4319-30, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21918195

ABSTRACT

The K/BxN serum transfer model of arthritis is critically dependent on FcγR signaling events mediated by spleen tyrosine kinase (Syk). However, the specific cell types in which this signaling is required are not known. We report that deletion of Syk in neutrophils, achieved using syk(f/f) MRP8-cre(+) mice, blocks disease development in serum transfer arthritis. The syk(f/f) MRP8-cre(+) mice display absent joint disease and reduced deposition of pathogenic anti-glucose-6-phosphate isomerase Abs in the joint (with a reciprocal accumulation of these Abs in the peripheral circulation). Additionally, syk(f/f) MRP8-cre(+) mice manifest poor edema formation within 3 h after formation of cutaneous immune complexes (Arthus reaction). Together, this suggests that neutrophil-dependent recognition of immune complexes contributes significantly to changes in vascular permeability during the early phases of immune complex disease. Using mixed chimeric mice, containing both wild-type and syk(f/f) MRP8-cre(+) neutrophils, we find no impairment in recruitment of Syk-deficient neutrophils to the inflamed joint, but they fail to become primed, demonstrating lower cytokine production after removal from the joint. They also display an increased apoptotic rate compared with wild-type cells in the same joint. Mast cell-deficient c-kit(sh/sh) mice developed robust arthritis after serum transfer whereas c-kit(W/Wv) mice did not, suggesting that previous conclusions concerning the central role of mast cells in this model may need to be revised. Basophil-deficient mice also responded normally to K/BxN serum transfer. These results demonstrate that Syk-dependent signaling in neutrophils alone is critically required for arthritis development in the serum transfer model.


Subject(s)
Arthritis, Experimental/metabolism , Arthus Reaction/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neutrophils/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction/immunology , Animals , Arthritis, Experimental/immunology , Arthus Reaction/immunology , Cell Separation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Intracellular Signaling Peptides and Proteins/immunology , Mast Cells/immunology , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Protein-Tyrosine Kinases/immunology , Syk Kinase
20.
Immunol Rev ; 228(1): 23-40, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19290919

ABSTRACT

Lyn is an Src family kinase present in B lymphocytes and myeloid cells. In these cell types, Lyn establishes signaling thresholds by acting as both a positive and a negative modulator of a variety of signaling responses and effector functions. Lyn deficiency in mice results in the development of myeloproliferation and autoimmunity. The latter has been attributed to the hyper-reactivity of Lyn-deficient B cells due to the unique role of Lyn in downmodulating B-cell receptor activation, mainly through phosphorylation of inhibitory molecules and receptors. Myeloproliferation results, on the other hand, from the enhanced sensitivity of Lyn-deficient progenitors to a number of colony-stimulating factors (CSFs). The hyper-sensitivity to myeloid growth factors may also be secondary to poor inhibitory receptor phosphorylation, leading to impaired recruitment/activation of tyrosine phosphatases and reduced downmodulation of CSF signaling responses. Despite these observations, the overall role of Lyn in the modulation of myeloid cell effector functions is much less well understood, as often both positive and negative roles of this kinase have been reported. In this review, we discuss the current knowledge of the duplicitous nature of Lyn in the modulation of myeloid cell signaling and function.


Subject(s)
Myeloid Cells/cytology , Myeloid Cells/metabolism , Signal Transduction , src-Family Kinases/metabolism , Animals , Blood Cells/metabolism , Humans , Immune System/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL