Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Immunol ; 199(11): 3914-3924, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29079698

ABSTRACT

Proteinase 3 (PR3) is a myeloid serine protease expressed in neutrophils, monocytes, and macrophages. PR3 has a number of well-characterized proinflammatory functions, including cleaving and activating chemokines and controlling cell survival and proliferation. When presented on the surface of apoptotic neutrophils, PR3 can disrupt the normal anti-inflammatory reprogramming of macrophages following the phagocytosis of apoptotic cells. To better understand the function of PR3 in vivo, we generated a human PR3 transgenic mouse (hPR3Tg). During zymosan-induced peritonitis, hPR3Tg displayed an increased accumulation of neutrophils within the peritoneal cavity compared with wild-type control mice, with no difference in the recruitment of macrophages or B or T lymphocytes. Mice were also subjected to cecum ligation and puncture, a model used to induce peritoneal inflammation through infection. hPR3Tg displayed decreased survival rates in acute sepsis, associated with increased neutrophil extravasation. The decreased survival and increased neutrophil accumulation were associated with the cleavage of annexin A1, a powerful anti-inflammatory protein known to facilitate the resolution of inflammation. Additionally, neutrophils from hPR3Tg displayed enhanced survival during apoptosis compared with controls, and this may also contribute to the increased accumulation observed during the later stages of inflammation. Taken together, our data suggest that human PR3 plays a proinflammatory role during acute inflammatory responses by affecting neutrophil accumulation, survival, and the resolution of inflammation.


Subject(s)
Myeloblastin/metabolism , Neutrophils/immunology , Peritoneal Cavity/pathology , Peritonitis/immunology , Sepsis/immunology , Animals , Annexin A1/metabolism , Apoptosis , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloblastin/genetics , Peritonitis/chemically induced , Phagocytosis , Sepsis/chemically induced , Zymosan
2.
J Biol Chem ; 291(20): 10476-89, 2016 May 13.
Article in English | MEDLINE | ID: mdl-26961880

ABSTRACT

Proteinase 3 (PR3), the autoantigen in granulomatosis with polyangiitis, is expressed at the plasma membrane of resting neutrophils, and this membrane expression increases during both activation and apoptosis. Using surface plasmon resonance and protein-lipid overlay assays, this study demonstrates that PR3 is a phosphatidylserine-binding protein and this interaction is dependent on the hydrophobic patch responsible for membrane anchorage. Molecular simulations suggest that PR3 interacts with phosphatidylserine via a small number of amino acids, which engage in long lasting interactions with the lipid heads. As phosphatidylserine is a major component of microvesicles (MVs), this study also examined the consequences of this interaction on MV production and function. PR3-expressing cells produced significantly fewer MVs during both activation and apoptosis, and this reduction was dependent on the ability of PR3 to associate with the membrane as mutating the hydrophobic patch restored MV production. Functionally, activation-evoked MVs from PR3-expressing cells induced a significantly larger respiratory burst in human neutrophils compared with control MVs. Conversely, MVs generated during apoptosis inhibited the basal respiratory burst in human neutrophils, and those generated from PR3-expressing cells hampered this inhibition. Given that membrane expression of PR3 is increased in patients with granulomatosis with polyangiitis, MVs generated from neutrophils expressing membrane PR3 may potentiate oxidative damage of endothelial cells and promote the systemic inflammation observed in this disease.


Subject(s)
Cell-Derived Microparticles/metabolism , Myeloblastin/metabolism , Phosphatidylserines/metabolism , Animals , Apoptosis , Cell Line , Granulomatosis with Polyangiitis/enzymology , Granulomatosis with Polyangiitis/etiology , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Myeloblastin/chemistry , Neutrophils/metabolism , Phospholipid Transfer Proteins/metabolism , Rats , Respiratory Burst
3.
Am J Respir Cell Mol Biol ; 54(5): 740-50, 2016 05.
Article in English | MEDLINE | ID: mdl-26517580

ABSTRACT

Neutrophil-associated inflammation during Pseudomonas aeruginosa lung infection is a determinant of morbidity in cystic fibrosis (CF). Neutrophil apoptosis is a key factor in inflammation resolution and is controlled by cytosolic proliferating cell nuclear antigen (PCNA). p21/Waf1, a cyclin-dependent kinase inhibitor, is a partner of PCNA, and its mRNA is up-regulated in human neutrophils during LPS challenge. We show here that, after 7 days of persistent infection with P. aeruginosa, neutrophilic inflammation was more prominent in p21(-/-) compared with wild-type (WT) mice. Notably, no intrinsic defect in the phagocytosis of apoptotic cells by macrophages was found in p21(-/-) compared with WT mice. Inflammatory cell analysis in peritoneal lavages after zymosan-induced peritonitis showed a significantly increased number of neutrophils at 48 hours in p21(-/-) compared with WT mice. In vitro analysis was consistent with delayed neutrophil apoptosis in p21(-/-) compared with WT mice. Ectopic expression of p21/waf1 in neutrophil-differentiated PLB985 cells potentiated apoptosis and reversed the prosurvival effect of PCNA. In human neutrophils, p21 messenger RNA was induced by TNF-α, granulocyte colony-stimulating factor, and LPS. Neutrophils isolated from patients with CF showed enhanced survival, which was reduced after treatment with a carboxy-peptide derived from the sequence of p21/waf1. Notably, p21/waf1 was detected by immunohistochemistry in neutrophils within lungs from patients with CF. Our data reveal a novel role for p21/waf1 in the resolution of inflammation via its ability to control neutrophil apoptosis. This mechanism may be relevant in the neutrophil-dominated inflammation observed in CF and other chronic inflammatory lung conditions.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Neutrophils/metabolism , Pneumonia/metabolism , Pneumonia/microbiology , Pseudomonas Infections/metabolism , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/physiology , Adolescent , Animals , Apoptosis/drug effects , Cell Count , Cell Differentiation/drug effects , Cell Line , Cyclin-Dependent Kinase Inhibitor p21/deficiency , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cystic Fibrosis/complications , Cystic Fibrosis/microbiology , Cystic Fibrosis/pathology , Female , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Models, Biological , Neutrophils/drug effects , Peritonitis/microbiology , Peritonitis/pathology , Phagocytosis/drug effects , Pneumonia/complications , Pneumonia/pathology , Proliferating Cell Nuclear Antigen/metabolism , Pseudomonas Infections/complications , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Zymosan
4.
Trends Immunol ; 32(3): 117-24, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21317039

ABSTRACT

Recently, unexpected biological features of polymorphonuclear neutrophils have been revealed. In addition to their pivotal role in the defence against pathogens, neutrophils display a high degree of plasticity and contribute to control of adaptive immune responses. An emerging aspect of neutrophils is their ability to modulate their survival in response to both intrinsic and extrinsic factors. This review focuses on recent advances that have uncovered proliferating cell nuclear antigen (PCNA) and other cell cycle regulatory proteins as novel players regulating neutrophil survival. A better understanding of the mechanisms involved in neutrophil fate might pave the way for the identification of new anti-inflammatory molecules.


Subject(s)
Apoptosis , Neutrophils/cytology , Neutrophils/immunology , Animals , Cell Nucleus/immunology , Cytoplasm/immunology , Humans , Inflammation/immunology , Proliferating Cell Nuclear Antigen/immunology
5.
J Immunol ; 189(5): 2574-83, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22844112

ABSTRACT

Proteinase 3 (PR3) is the target of anti-neutrophil cytoplasm Abs in granulomatosis with polyangiitis, a form of systemic vasculitis. Upon neutrophil apoptosis, PR3 is coexternalized with phosphatidylserine and impaired macrophage phagocytosis. Calreticulin (CRT), a protein involved in apoptotic cell recognition, was found to be a new PR3 partner coexpressed with PR3 on the neutrophil plasma membrane during apoptosis, but not after degranulation. The association between PR3 and CRT was demonstrated in neutrophils by confocal microscopy and coimmunoprecipitation. Evidence for a direct interaction between PR3 and the globular domain of CRT, but not with its P domain, was provided by surface plasmon resonance spectroscopy. Phagocytosis of apoptotic neutrophils from healthy donors was decreased after blocking lipoprotein receptor-related protein (LRP), a CRT receptor on macrophages. In contrast, neutrophils from patients with granulomatosis with polyangiitis expressing high membrane PR3 levels showed a lower rate of phagocytosis than those from healthy controls not affected by anti-LRP, suggesting that the LRP-CRT pathway was disturbed by PR3-CRT association. Moreover, phagocytosis of apoptotic PR3-expressing cells potentiated proinflammatory cytokine in vitro by human monocyte-derived macrophages and in vivo by resident murine peritoneal macrophages, and diverted the anti-inflammatory response triggered by the phagocytosis of apoptotic cells after LPS challenge in thioglycolate-elicited murine macrophages. Therefore, membrane PR3 expressed on apoptotic neutrophils might amplify inflammation and promote autoimmunity by affecting the anti-inflammatory "reprogramming" of macrophages.


Subject(s)
Apoptosis/immunology , Autoantigens/metabolism , Calreticulin/metabolism , Granulomatosis with Polyangiitis/immunology , Macrophages/immunology , Microscopic Polyangiitis/immunology , Myeloblastin/metabolism , Neutrophils/immunology , Adjuvants, Immunologic/physiology , Animals , Granulomatosis with Polyangiitis/enzymology , Granulomatosis with Polyangiitis/pathology , Humans , Inflammation/enzymology , Inflammation/immunology , Inflammation/pathology , Macrophages/enzymology , Macrophages/pathology , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Microscopic Polyangiitis/enzymology , Microscopic Polyangiitis/pathology , Neutrophils/enzymology , Neutrophils/pathology , Rats
6.
Postgrad Med J ; 90(1063): 290-6, 2014 May.
Article in English | MEDLINE | ID: mdl-24737903

ABSTRACT

Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitides are a heterogeneous group of diseases corresponding to necrotising inflammation of small vessels with a wide range of clinical presentations. At least two of the diseases are believed to exhibit a common ground of pathophysiological mechanisms. These are granulomatosis with polyangiitis (GPA, formerly known as Wegener's granulomatosis) and microscopic polyangiitis (MPA). ANCA directed against proteinase 3 (PR3) are preferentially associated with GPA, and anti-myeloperoxidase (MPO) ANCA are associated mainly with MPA and eosinophilic GPA (formerly known as Churg-Strauss syndrome). Anti-MPO and anti-PR3 antibodies can activate neutrophils in vitro. In vivo data are available for humans and mice on the pathogenicity of anti-MPO but it is more controversial for PR3-ANCA. A recent genome-wide association study of patients with ANCA-associated vasculitides confirmed the genetic contribution to the pathogenesis of these conditions, with significant association of PR3-ANCA and human leukocyte antigen-DP and the genes encoding α1-antitrypsin and PR3. MPO-ANCA were significantly associated with human leukocyte antigen-DQ. Thus, recent results from epidemiological studies, genome-wide association study and therapeutic trials have suggested that these entities are, in fact, distinct. We have summarised these results and discuss the idea that these two entities should be studied separately as the nature of the two auto-antigens suggests at a molecular level despite shared ANCA involvement.


Subject(s)
Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/immunology , Granulomatosis with Polyangiitis/immunology , Microscopic Polyangiitis/immunology , alpha 1-Antitrypsin/immunology , Animals , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/genetics , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/pathology , Antibodies, Antineutrophil Cytoplasmic/immunology , Autoantigens , Gene-Environment Interaction , Genetic Predisposition to Disease , Genome-Wide Association Study , Granulomatosis with Polyangiitis/genetics , Granulomatosis with Polyangiitis/pathology , Humans , Mice , Microscopic Polyangiitis/genetics , Microscopic Polyangiitis/pathology , Polymorphism, Single Nucleotide , alpha 1-Antitrypsin/genetics
7.
Mol Cancer Ther ; 23(4): 421-435, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38030380

ABSTRACT

IL12 is a proinflammatory cytokine, that has shown promising antitumor activity in humans by promoting the recruitment and activation of immune cells in tumors. However, the systemic administration of IL12 has been accompanied by considerable toxicity, prompting interest in researching alternatives to drive preferential IL12 bioactivity in the tumor. Here, we have generated XTX301, a tumor-activated IL12 linked to the human Fc protein via a protease cleavable linker that is pharmacologically inactivated by an IL12 receptor subunit beta 2 masking domain. In vitro characterization demonstrates multiple matrix metalloproteases, as well as human primary tumors cultured as cell suspensions, can effectively activate XTX301. Intravenous administration of a mouse surrogate mXTX301 demonstrated significant tumor growth inhibition (TGI) in inflamed and non-inflamed mouse models without causing systemic toxicities. The superiority of mXTX301 in mediating TGI compared with non-activatable control molecules and the greater percentage of active mXTX301 in tumors versus other organs further confirms activation by the tumor microenvironment-associated proteases in vivo. Pharmacodynamic characterization shows tumor selective increases in inflammation and upregulation of immune-related genes involved in IFNγ cell signaling, antigen processing, presentation, and adaptive immune response. XTX301 was tolerated following four repeat doses up to 2.0 mg/kg in a nonhuman primate study; XTX301 exposures were substantially higher than those at the minimally efficacious dose in mice. Thus, XTX301 has the potential to achieve potent antitumor activity while widening the therapeutic index of IL12 treatment and is currently being evaluated in a phase I clinical trial.


Subject(s)
Interleukin-12 , Neoplasms , Humans , Mice , Animals , Interleukin-12/metabolism , Neoplasms/drug therapy , Cytokines , Signal Transduction , Therapeutic Index , Tumor Microenvironment
8.
J Biol Chem ; 287(40): 33812-25, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22846997

ABSTRACT

Neutrophils are deprived of proliferative capacity and have a tightly controlled lifespan to avoid their persistence at the site of injury. We have recently described that the proliferating cell nuclear antigen (PCNA), a nuclear factor involved in DNA replication and repair of proliferating cells, is a key regulator of neutrophil survival. In neutrophils, PCNA was localized exclusively in the cytoplasm due to its nuclear-to-cytoplasmic relocalization during granulocytic differentiation. We showed here that leptomycin B, an inhibitor of the chromosome region maintenance 1 (CRM1) exportin, inhibited PCNA relocalization during granulocytic differentiation of HL-60 and NB4 promyelocytic cell lines and of human CD34(+) primary cells. Using enhanced green fluorescent protein fusion constructs, we have demonstrated that PCNA relocalization involved a nuclear export signal (NES) located from Ile-11 to Ile-23 in the PCNA sequence. However, this NES, located at the inner face of the PCNA trimer, was not functional in wild-type PCNA, but instead, was fully active and leptomycin B-sensitive in the monomeric PCNAY114A mutant. To test whether a defect in PCNA cytoplasmic relocalization would affect its antiapoptotic activity in mature neutrophils, a chimeric PCNA fused with the SV40 nuclear localization sequence (NLS) was generated to preclude its cytoplasmic localization. As expected, neutrophil-differentiated PLB985 cells expressing ectopic SV40NLS-PCNA had an increased nuclear PCNA as compared with cells expressing wild-type PCNA. Accordingly, the nuclear PCNA mutant did not show any antiapoptotic activity as compared with wild-type PCNA. Nuclear-to-cytoplasmic relocalization that occurred during myeloid differentiation is essential for PCNA antiapoptotic activity in mature neutrophils and is dependent on the newly identified monomerization-dependent PCNA NES.


Subject(s)
Apoptosis , Cell Nucleus/metabolism , Cytoplasm/metabolism , Karyopherins/metabolism , Neutrophils/cytology , Proliferating Cell Nuclear Antigen/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Active Transport, Cell Nucleus , Blotting, Western , Cell Differentiation , Cells, Cultured , Fatty Acids, Unsaturated/chemistry , Granulocytes/cytology , HL-60 Cells , HeLa Cells , Humans , Inflammation , Models, Molecular , Mutation , Neutropenia/metabolism , Exportin 1 Protein
9.
Ann Rheum Dis ; 72(8): 1273-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23606701

ABSTRACT

Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitides are a heterogeneous group of diseases corresponding to necrotising inflammation of small vessels with a wide range of clinical presentations. At least two of the diseases are believed to exhibit a common ground of pathophysiological mechanisms. These are granulomatosis with polyangiitis (GPA, formerly known as Wegener's granulomatosis) and microscopic polyangiitis (MPA). ANCA directed against proteinase 3 (PR3) are preferentially associated with GPA, and anti-myeloperoxidase (MPO) ANCA are associated mainly with MPA and eosinophilic GPA (formerly known as Churg-Strauss syndrome). Anti-MPO and anti-PR3 antibodies can activate neutrophils in vitro. In vivo data are available for humans and mice on the pathogenicity of anti-MPO but it is more controversial for PR3-ANCA. A recent genome-wide association study of patients with ANCA-associated vasculitides confirmed the genetic contribution to the pathogenesis of these conditions, with significant association of PR3-ANCA and human leukocyte antigen-DP and the genes encoding α1-antitrypsin and PR3. MPO-ANCA were significantly associated with human leukocyte antigen-DQ. Thus, recent results from epidemiological studies, genome-wide association study and therapeutic trials have suggested that these entities are, in fact, distinct. We have summarised these results and discuss the idea that these two entities should be studied separately as the nature of the two auto-antigens suggests at a molecular level despite shared ANCA involvement.


Subject(s)
Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/classification , Terminology as Topic , Animals , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/immunology , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/physiopathology , Antibodies, Antineutrophil Cytoplasmic/immunology , Autoantigens/genetics , Autoantigens/immunology , Churg-Strauss Syndrome/classification , Churg-Strauss Syndrome/immunology , Churg-Strauss Syndrome/physiopathology , Genome-Wide Association Study , Granulomatosis with Polyangiitis/classification , Granulomatosis with Polyangiitis/immunology , Granulomatosis with Polyangiitis/physiopathology , Humans , Mice , Microscopic Polyangiitis/classification , Microscopic Polyangiitis/immunology , Microscopic Polyangiitis/physiopathology , Myeloblastin/genetics , Myeloblastin/immunology , Peroxidase/immunology , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/immunology
10.
J Immunother Cancer ; 11(12)2023 12 12.
Article in English | MEDLINE | ID: mdl-38164757

ABSTRACT

INTRODUCTION: The clinical benefit of the anti-CTLA-4 monoclonal antibody (mAb) ipilimumab has been well established but limited by immune-related adverse events, especially when ipilimumab is used in combination with anti-PD-(L)1 mAb therapy. To overcome these limitations, we have developed XTX101, a tumor-activated, Fc-enhanced anti-CTLA-4 mAb. METHODS: XTX101 consists of an anti-human CTLA-4 mAb covalently linked to masking peptides that block the complementarity-determining regions, thereby minimizing the mAb binding to CTLA-4. The masking peptides are designed to be released by proteases that are typically dysregulated within the tumor microenvironment (TME), resulting in activation of XTX101 intratumorally. Mutations within the Fc region of XTX101 were included to enhance affinity for FcγRIII, which is expected to enhance potency through antibody-dependent cellular cytotoxicity. RESULTS: Biophysical, biochemical, and cell-based assays demonstrate that the function of XTX101 depends on proteolytic activation. In human CTLA-4 transgenic mice, XTX101 monotherapy demonstrated significant tumor growth inhibition (TGI) including complete responses, increased intratumoral CD8+T cells, and regulatory T cell depletion within the TME while maintaining minimal pharmacodynamic effects in the periphery. XTX101 in combination with anti-PD-1 mAb treatment resulted in significant TGI and was well tolerated in mice. XTX101 was activated in primary human tumors across a range of tumor types including melanoma, renal cell carcinoma, colon cancer and lung cancer in an ex vivo assay system. CONCLUSIONS: These data demonstrate that XTX101 retains the full potency of an Fc-enhanced CTLA-4 antagonist within the TME while minimizing the activity in non-tumor tissue, supporting the further evaluation of XTX101 in clinical studies.


Subject(s)
Antineoplastic Agents , Melanoma , Humans , Mice , Animals , CTLA-4 Antigen , Ipilimumab/therapeutic use , Antineoplastic Agents/therapeutic use , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Melanoma/drug therapy , Disease Models, Animal , Mice, Transgenic , Peptides/therapeutic use , Tumor Microenvironment
11.
Blood ; 116(20): 4288-96, 2010 Nov 18.
Article in English | MEDLINE | ID: mdl-20705760

ABSTRACT

Human polymorphonuclear leukocytes adhesion to endothelial cells during the early stage of inflammation leads to cell surface externalization of Annexin A1 (AnxA1), an effector of endogenous anti-inflammation. The antiadhesive properties of AnxA1 become operative to finely tune polymorphonuclear leukocytes transmigration to the site of inflammation. Membrane bound proteinase 3 (PR3) plays a key role in this microenvironment by cleaving the N terminus bioactive domain of AnxA1. In the present study, we generated a PR3-resistant human recombinant AnxA1-named superAnxA1 (SAnxA1)-and tested its in vitro and in vivo properties in comparison to the parental protein. SAnxA1 bound and activated formyl peptide receptor 2 in a similar way as the parental protein, while showing a resistance to cleavage by recombinant PR3. SAnxA1 retained anti-inflammatory activities in the murine inflamed microcirculation (leukocyte adhesion being the readout) and in skin trafficking model. When longer-lasting models of inflammation were applied, SAnxA1 displayed stronger anti-inflammatory effect over time compared with the parental protein. Together these results indicate that AnxA1 cleavage is an important process during neutrophilic inflammation and that controlling the balance between AnxA1/PR3 activities might represent a promising avenue for the discovery of novel therapeutic approaches.


Subject(s)
Annexin A1/metabolism , Inflammation/pathology , Inflammation/prevention & control , Microvessels/pathology , Mutant Proteins/metabolism , Protein Engineering , Amino Acid Sequence , Animals , Annexin A1/chemistry , Annexin A1/isolation & purification , Anti-Inflammatory Agents/metabolism , Cell Adhesion , Cell Communication , Cell Movement , Endothelial Cells/metabolism , Female , HEK293 Cells , Humans , Male , Mice , Microvessels/metabolism , Molecular Sequence Data , Mutant Proteins/chemistry , Neutrophils/cytology , Neutrophils/metabolism , Protein Binding , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
12.
Exp Hematol ; 88: 15-27, 2020 08.
Article in English | MEDLINE | ID: mdl-32721504

ABSTRACT

Thrombopoietin (TPO), through activation of its cognate receptor Mpl, is the major regulator of platelet production. However, residual platelets observed in TPO- and Mpl-loss-of-function (LOF) mice suggest the existence of an additional factor to TPO in platelet production. As erythropoietin (EPO) exhibited both in vitro megakaryocytic potential, in association with other early-acting cytokines, and in vivo platelet activation activity, we sought to investigate its role in this setting. Here, we used multiple LOF models to decipher the reciprocal role of EPO and TPO in the regulation of platelet production in TPO-LOF and Mpl-LOF mice and of platelet size heterogeneity in wild-type mice. We first identified EPO as the major thrombopoietic factor in the absence of the TPO-Mpl pathway. Based on the study of several mouse models we found that the EPO-EPO receptor pathway acts on late-stage megakaryopoiesis and is responsible for large-sized platelet production, while the TPO-Mpl pathway promotes small-sized platelet production. On the basis of our data, EPO might be used for thrombocytopenia supportive therapy in congenital amegakaryocytopoiesis. Furthermore, as a distribution skewed toward large platelets is an independent risk factor and a poor prognosis indicator in atherothrombosis, the characterization of EPO's role in the production of large-sized platelets, if confirmed in humans, may open new perspectives in the understanding of the role of EPO-induced platelets in atherothrombosis.


Subject(s)
Blood Platelets/metabolism , Erythropoietin/metabolism , Megakaryocytes/microbiology , Thrombopoiesis , Thrombopoietin/metabolism , Animals , Erythropoietin/genetics , Female , Mice , Mice, Knockout , Thrombopoietin/genetics
13.
Contrib Microbiol ; 15: 118-146, 2008.
Article in English | MEDLINE | ID: mdl-18511859

ABSTRACT

Polymorphonuclear neutrophils (PMNs) and monocyte/macrophages (MMs) are professional phagocytic cells that are able to phagocytose and destroy infectious agents. Therefore, they are key anti-infectious actors in host defense but can mediate tissue damages. In addition, it is now clear that the role of these cells goes far beyond phagocytosis and pathogen killing. PMNs and MMs are essential cells for immunity, absolutely required to build and modulate the innate response. The respective roles of PMNs and MMs in the inflammatory process are discussed: their common features and their differences are reviewed, both in terms of origins and functions with special emphasis on novel concepts about neutrophil survival and resolution of inflammation. The recognition and the subsequent engulfment of apoptotic PMNs by macrophages is a key event of the resolution of inflammation, which can be associated with autoimmunity or inflammatory diseases. During the past years, significant efforts have been made to dissect the molecular mechanisms governing phagocytosis and pathogen killing. Although these effector functions are crucial, more work has to be done to understand the respective role of PMNs and MMs to regulate and inhibit the inflammatory process as well as the immune response. This might be the future challenge for the next years in phagocyte research and this will presumably open new avenues of research in the modulation of inflammation.


Subject(s)
Immunity, Innate , Monocytes/immunology , Neutrophils/immunology , Animals , Apoptosis , Cell Adhesion , Cell Movement , Humans , Inflammation/immunology , Macrophages/immunology , Macrophages/physiology , Monocytes/physiology , Neutrophils/physiology , Receptors, Immunologic/immunology , Signal Transduction
14.
J Leukoc Biol ; 94(4): 723-31, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23825390

ABSTRACT

We have shown previously that PCNA, a nuclear factor involved in DNA replication and repair in proliferating cells, is localized exclusively in the cytoplasm of neutrophils, where it regulates their survival. Nuclear PCNA functions are tightly linked to its ring-shaped structure, which allows PCNA to bind to numerous partner proteins to orchestrate DNA-related processes. We have shown that only monomeric PCNA can expose its NES to be relocalized from nucleus to cytosol during granulocyte differentiation. This study tested the hypothesis that monomeric PCNA could have a biological role in neutrophils. With the use of a combination of cross-linking and gel-filtration experiments, trimeric and monomeric PCNAs were detected in neutrophil cytosol. The promyelocytic cell line PLB985 was next stably transfected to express the monomeric PCNAY114A mutant to examine its function compared with the WT trimeric PCNA. Monomeric PCNAY114A mutant potentiated DMF-induced differentiation, as evidenced by an increased percentage of CD11b- and gp91phox-positive PLB985PCNAY114A cells and by an increased, opsonized zymosan-triggered NADPH oxidase activity compared with PLB985PCNA or PLB985 cells overexpressing WT PCNA or the empty plasmid, respectively. Regarding antiapoptotic activity, DMF-differentiated PLB985 cells overexpressing WT or the monomeric PCNAY114A mutant displayed a similar antiapoptotic activity following treatment with gliotoxin or TRAIL compared with PLB985. The molecular basis through which cytoplasmic PCNA exerts its antiapoptotic activity in mature neutrophils may, at least in part, be independent of the trimeric conformation.


Subject(s)
Cytosol/metabolism , Neutrophils/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Apoptosis/drug effects , CD11b Antigen/metabolism , Cell Differentiation/drug effects , Cell Survival/drug effects , Chromatography, Gel , Cytosol/drug effects , Dimethylformamide/pharmacology , HeLa Cells , Humans , Membrane Glycoproteins/metabolism , Mutant Proteins/metabolism , NADPH Oxidase 2 , NADPH Oxidases/metabolism , Neutrophils/cytology , Neutrophils/drug effects , Neutrophils/enzymology , Proliferating Cell Nuclear Antigen/chemistry , Protein Multimerization/drug effects , Tetradecanoylphorbol Acetate/pharmacology
15.
Front Immunol ; 3: 311, 2012.
Article in English | MEDLINE | ID: mdl-23181059

ABSTRACT

New therapeutic approaches that can accelerate neutrophil apoptosis under inflammatory conditions to enhance the resolution of inflammation are now under study. Neutrophils are deprived of proliferative capacity and have a tightly controlled lifespan to avoid their persistence at the site of injury. We have recently described that the proliferating cell nuclear antigen (PCNA), a nuclear factor involved in DNA replication and repair of proliferating cells is a key regulator of neutrophil survival. The nuclear-to-cytoplasmic relocalization occurred during granulocytic differentiation and is dependent on a nuclear export sequence thus strongly suggesting that PCNA has physiologic cytoplasmic functions. In this review, we will try to put into perspective the physiologic relevance of PCNA in neutrophils. We will discuss key issues such as molecular structure, post-translational modifications, based on our knowledge of nuclear PCNA, assuming that similar principles governing its function are conserved between nuclear and cytosolic PCNA. The example of cystic fibrosis that features one of the most intense neutrophil-dominated pulmonary inflammation will be discussed. We believe that through an intimate comprehension of the cytosolic PCNA scaffold based on nuclear PCNA knowledge, novel pathways regulating neutrophil survival can be unraveled and innovative agents can be developed to dampen inflammation where it proves detrimental.

16.
Presse Med ; 41(10): 996-1003, 2012 Oct.
Article in French | MEDLINE | ID: mdl-22921085

ABSTRACT

ANCA-associated vasculitides comprise granulomatosis with polyangiitis (GPA) (formerly names Wegener's granulomatosis), Churg-Strauss syndrome (SCS) (which will be renamed GPA and eosinophilia) and microscopic polyangiitis (MPA). Immune cells (dendritic and non dendritic cells) and inflammatory cells (neutrophils, monocytes, macrophages) and resident cells (endothelial cells, fibroblasts) are implicated in the pathophysiology of ANCA-associated vasculitides. One of the targets of ANCA, myeloperoxydase, is only present in the azurophil granules of neutrophils, whereas the other target of these antibodies, proteinase 3, is also present at the internal face of cytoplasmic membrane of neutrophils, as well as at their surface. Anti-myeloperoxydase ANCA are pathogenicin vitroandin vivo, whereas the pathogenicity of anti-proteinase 3 ANCA has only been demonstrated in vitro and recent studies suggest a pathogenic role of ANCA anti-PR3 in mouse model. Two phenotypes of GPA can be distinguished: a granulomatous form, localized to the respiratory tract with Th1 immune response features, and a vasculitic form with Th2 immune response features. Recently, an increase in TH17 lymphocytes at the acute phase and a defect in T regulatory cells at the chronic phase have been identified in GPA. The role of B-lymphocytes in the pathogenesis of ANCA-associated vasculitides is now well documented by the effectiveness of rituximab in the treatment of this condition.


Subject(s)
Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/etiology , Antibodies, Antineutrophil Cytoplasmic/physiology , Animals , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/genetics , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/immunology , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/pathology , Antibodies, Antineutrophil Cytoplasmic/genetics , Antibodies, Antineutrophil Cytoplasmic/immunology , Eosinophils/immunology , Eosinophils/pathology , Eosinophils/physiology , Genetic Predisposition to Disease , Humans , Mice , Models, Biological
17.
Int Immunopharmacol ; 11(1): 55-66, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20974309

ABSTRACT

Recent studies have associated the dysregulated expression of Annexin-A1/Formyl peptide receptor 2 (FPR2/ALX) system with the development of autoimmune diseases. In this study we systematically scanned human leukocyte subsets for the presence of this pathway aiming to provide a roadmap that will help investigators to explore possible links between the development of immune related disorders and the expression of this system. Our results show that neutrophils, monocytes and NK cells express higher levels of both AnxA1 and FPR2/ALX compared to T or B cells. Further analysis of specific T cell subsets revealed higher levels in activated CD25(+) and memory CD45RO CD4 T cells compared to resting CD25(-) or naïve CD45RA CD4 T cells. Together the results expand our knowledge of the AnxA1-FPR2/ALX system in immune cells and provide new avenues for investigation into the functions of this signalling pathway in systems other than that classically described for neutrophils.


Subject(s)
Annexin A1/biosynthesis , Leukocytes/immunology , Leukocytes/metabolism , Receptors, Formyl Peptide/biosynthesis , Receptors, Lipoxin/biosynthesis , Adult , Autoimmune Diseases/metabolism , Blotting, Western , Female , Flow Cytometry , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Neutrophils/immunology , Neutrophils/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Young Adult
18.
Biochem Pharmacol ; 79(4): 525-34, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19765545

ABSTRACT

The study of the inflammatory response has seen a tremendous expansion over the last 30 years. Advancements in technology and better knowledge of the ethiopathogenesis of several inflammatory conditions have facilitated this process allowing researchers to almost reach the core of problem. Thus, we now know that inflammation can be manifested in many different ways depending on the context that has elicited it. Viral and infectious, allergic and autoimmune, carcinogenic and resolutive are just a few examples of how inflammation can disguise itself. However, and most intriguingly, it appears that the more we try to discover "an ideal target" and delineate borders for a specific class of inflammatory conditions the more we find similarities, overlaps or often links that we did not predict. These somehow disappointing findings have pushed researchers towards a frantic search for new and more "reliable" targets. As result, we have recently seen a surge of many novel mediators of inflammation. If we just limit our focus to inflammatory cytokines, the main topic of this commentary, the list seems never-ending: IL-15, IL-17, IL-18, IL-21, IL-22, IL-23, IL-27 and IL-33. Are these cytokines destined to supersede prostaglandins and other autacoids for their key role in inflammation? Are we going to see a cheap and effective alternative to aspirin on the supermarket shelves in the next few years? Here we summarize the most recent findings on the biological effects of these new inflammatory cytokines and discuss how these discoveries might influence our current view on therapeutic approaches to treat inflammation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Aspirin/therapeutic use , Drug Delivery Systems/trends , Inflammation Mediators/therapeutic use , Interleukin-15/physiology , Interleukins/physiology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Drug Delivery Systems/methods , Humans , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Inflammation/virology , Inflammation Mediators/physiology , Interleukin-33
19.
J Exp Med ; 207(12): 2631-45, 2010 Nov 22.
Article in English | MEDLINE | ID: mdl-20975039

ABSTRACT

Neutrophil apoptosis is a highly regulated process essential for inflammation resolution, the molecular mechanisms of which are only partially elucidated. In this study, we describe a survival pathway controlled by proliferating cell nuclear antigen (PCNA), a nuclear factor involved in DNA replication and repairing of proliferating cells. We show that mature neutrophils, despite their inability to proliferate, express high levels of PCNA exclusively in their cytosol and constitutively associated with procaspases, presumably to prevent their activation. Notably, cytosolic PCNA abundance decreased during apoptosis, and increased during in vitro and in vivo exposure to the survival factor granulocyte colony-stimulating factor (G-CSF). Peptides derived from the cyclin-dependent kinase inhibitor p21, which compete with procaspases to bind PCNA, triggered neutrophil apoptosis thus demonstrating that specific modification of PCNA protein interactions affects neutrophil survival. Furthermore, PCNA overexpression rendered neutrophil-differentiated PLB985 myeloid cells significantly more resistant to TNF-related apoptosis-inducing ligand- or gliotoxin-induced apoptosis. Conversely, a decrease in PCNA expression after PCNA small interfering RNA transfection sensitized these cells to apoptosis. Finally, a mutation in the PCNA interdomain-connecting loop, the binding site for many partners, significantly decreased the PCNA-mediated antiapoptotic effect. These results identify PCNA as a regulator of neutrophil lifespan, thereby highlighting a novel target to potentially modulate pathological inflammation.


Subject(s)
Neutrophils/physiology , Proliferating Cell Nuclear Antigen/physiology , Apoptosis , Caspase 3/physiology , Caspase 9/physiology , Cell Differentiation , Cell Nucleus/chemistry , Cell Survival , Cyclin-Dependent Kinase Inhibitor p21/physiology , Cytoplasm/chemistry , Humans , Peptide Fragments/physiology , Proliferating Cell Nuclear Antigen/analysis , RNA, Small Interfering/genetics
20.
Blood ; 110(12): 4086-95, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17712045

ABSTRACT

Proteinase 3 (PR3), a serine proteinase contained in neutrophil azurophilic granules, is considered a risk factor for vasculitides and rheumatoid arthritis when expressed on the outer leaflet of neutrophil plasma membrane and is the preferred target of antineutrophil cytoplasm autoantibodies (ANCA) in Wegener granulomatosis. ANCA binding to PR3 expressed at the surface of neutrophils activates them. Evidence is provided that neutrophil apoptosis induced significantly more membrane PR3 expression without degranulation (but no enhanced membrane CD35, CD66b, CD63, myeloperoxidase, or elastase expression). This observation was confirmed on cytoplasts, a model of granule-free neutrophils. We hypothesized that PR3 could interact with proteins involved in membrane flip-flop (eg, phospholipid scramblase 1 [PLSCR1]). PR3-PLSCR1 interaction in neutrophils was demonstrated by confocal microscopy and coimmunoprecipitation. In the RBL-2H3 rat mast-cell line stably transfected with PR3 or its inactive mutant (PR3S203A), PR3 externalization depended on PLSCR1, as shown by less PR3 externalization in the presence of rPLSCR1 siRNA, but independently of its serine-proteinase activity. Finally, apoptosis-externalized PR3 decreased the human macrophage-phagocytosis rate of apoptotic PR3 transfectants. Therefore, in addition to ANCA binding in vasculitis, the proinflammatory role of membrane PR3 expression may involve interference with macrophage clearance of apoptotic neutrophils.


Subject(s)
Apoptosis , Macrophages/enzymology , Myeloblastin/metabolism , Neutrophils/enzymology , Phagocytosis , Phospholipid Transfer Proteins/metabolism , Animals , Antibodies, Antineutrophil Cytoplasmic/immunology , Antibodies, Antineutrophil Cytoplasmic/metabolism , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , Apoptosis/genetics , Apoptosis/immunology , Arthritis, Rheumatoid/enzymology , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Cell Line , Cell Membrane/enzymology , Cell Membrane/genetics , Cell Membrane/immunology , Gene Expression Regulation, Enzymologic/immunology , Granulomatosis with Polyangiitis/enzymology , Granulomatosis with Polyangiitis/genetics , Granulomatosis with Polyangiitis/immunology , Humans , Macrophages/immunology , Mast Cells/enzymology , Mast Cells/immunology , Mutation/immunology , Myeloblastin/genetics , Myeloblastin/immunology , Neutrophil Activation/genetics , Neutrophil Activation/immunology , Neutrophils/immunology , Neutrophils/metabolism , Pancreatic Elastase/genetics , Pancreatic Elastase/immunology , Pancreatic Elastase/metabolism , Peroxidase/genetics , Peroxidase/immunology , Peroxidase/metabolism , Phagocytosis/genetics , Phagocytosis/immunology , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/immunology , Protein Transport/genetics , Protein Transport/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Rats , Risk Factors , Secretory Vesicles/enzymology , Secretory Vesicles/genetics , Secretory Vesicles/immunology , Vasculitis/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL