Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Rheumatology (Oxford) ; 60(7): 3388-3397, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33351137

ABSTRACT

OBJECTIVES: Circulating anti-ENO1 and anti-H2A IgG2 have been identified as specific signatures of LN in a cross-over approach. We sought to show whether the same antibodies identify selected population of patients with LN with potentially different clinical outcomes. METHODS: Here we report the prospective analysis over 36 months of circulating IgG2 levels in patients with newly diagnosed LN (n=91) and SLE (n=31) and in other patients with SLE recruited within 2 years from diagnosis (n=99). Anti-podocyte (ENO1), anti-nucleosome (DNA, histone 2 A, histone 3) and anti-circulating proteins (C1q, AnnexinA1-ANXA1) IgG2 antibodies were determined by home-made techniques. RESULTS: LN patients were the main focus of the study. Anti-ENO1, anti-H2A and anti-ANXA1 IgG2 decreased in parallel to proteinuria and normalized within 12 months in the majority of patients while anti-dsDNA IgG2 remained high over the 36 months. Anti-ENO1 and anti-H2A had the highest association with proteinuria (Heat Map) and identified the highest number of patients with high proteinuria (68% and 71% respectively) and/or with reduced estimated glomerula filtration rate (eGFR) (58% for both antibodies) compared with 23% and 17% of anti-dsDNA (agreement analysis). Anti-ENO1 positive LN patients had higher proteinuria than negative patients at T0 and presented the maximal decrement within 12 months. CONCLUSIONS: Anti-ENO1, anti-H2A and anti-ANXA1 antibodies were associated with high proteinuria in LN patients and Anti-ENO1 also presented the maximal reduction within 12 months that paralleled the decrease of proteinuria. Anti-dsDNA were not associated with renal outcome parameters. New IgG2 antibody signatures should be utilized as tracers of personalized therapies in LN. TRIAL REGISTRATION: The Zeus study was registered at https://clinicaltrials.gov (study number: NCT02403115).


Subject(s)
Immunoglobulin G/immunology , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/immunology , Adult , Annexin A1/immunology , Antibodies, Antinuclear/immunology , Autoantibodies/immunology , Biomarkers, Tumor/immunology , Complement C1q/immunology , DNA/immunology , DNA-Binding Proteins/immunology , Disease Progression , Female , Histones/immunology , Humans , Male , Middle Aged , Nucleosomes/immunology , Phosphopyruvate Hydratase/immunology , Prospective Studies , Tumor Suppressor Proteins/immunology
2.
Rheumatology (Oxford) ; 60(7): 3176-3188, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33374003

ABSTRACT

OBJECTIVES: Serum anti-dsDNA and anti-nucleosome IgGs have been proposed as signatures for SLE and LN in limited numbers of patients. We sought to show higher sensitivity and specificity of the same antibodies with the IgG2 isotype and included IgG2 antibodies vs specific intracellular antigens in the analysis. METHODS: A total of 1052 SLE patients with (n = 479) and without (n = 573) LN, recruited at different times from the beginning of symptoms, were included in the study. Patients with primary APS (PAPS, n = 24), RA (RA, n = 24) and UCTD (UCTD, n = 96) were analysed for comparison. Anti-nucleosome (dsDNA, Histone2A, Histone3), anti-intracellular antigens (ENO1), anti-annexin A1 and anti-C1q IgG2 were determined by non-commercial techniques. RESULTS: The presence in the serum of the IgG2 panel was highly discriminatory for SLE/LN vs healthy subjects. Serum levels of anti-dsDNA and anti-C1q IgG2 were more sensitive than those of IgGs (Farr radioimmunoassay/commercial assays) in identifying SLE patients at low-medium increments. Of more importance, serum positivity for anti-ENO1 and anti-H2A IgG2 discriminated between LN and SLE (ROC T0-12 months), and high levels at T0-1 month were detected in 63% and 67%, respectively, of LN, vs 3% and 3%, respectively, of SLE patients; serum positivity for each of these was correlated with high SLEDAI values. Minor differences existed between LN/SLE and the other rheumatologic conditions. CONCLUSION: Nephritogenic IgG2 antibodies represent a specific signature of SLE/LN, with a few overlaps with other rheumatologic conditions. High levels of anti-ENO1 and anti-H2A IgG2 correlated with SLE activity indexes and were discriminatory between SLE patients limited to the renal complication and other SLE patients. TRIAL REGISTRATION: The Zeus study was registered at https://clinicaltrials.gov, NCT02403115.


Subject(s)
Antibodies, Antinuclear/immunology , Immunoglobulin G/immunology , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/immunology , Adolescent , Adult , Annexin A1/immunology , Antibody Specificity , Antiphospholipid Syndrome/immunology , Arthritis, Rheumatoid/immunology , Biomarkers, Tumor/immunology , Complement C1q/immunology , Cross-Sectional Studies , DNA/immunology , DNA-Binding Proteins/immunology , Female , Histones/immunology , Humans , Male , Middle Aged , Nucleosomes/immunology , Phosphopyruvate Hydratase/immunology , Tumor Suppressor Proteins/immunology , Undifferentiated Connective Tissue Diseases/immunology , Young Adult
3.
J Immunol ; 203(7): 1753-1765, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31462505

ABSTRACT

Chronic neuroinflammation is a key pathological hallmark of multiple sclerosis (MS) that suggests that resolution of inflammation by specialized proresolving molecules is dysregulated in the disease. Annexin A1 (ANXA1) is a protein induced by glucocorticoids that facilitates resolution of inflammation through several mechanisms that include an inhibition of leukocyte recruitment and activation. In this study, we investigated the ability of ANXA1 to influence T cell effector function in relapsing/remitting MS (RRMS), an autoimmune disease sustained by proinflammatory Th1/Th17 cells. Circulating expression levels of ANXA1 in naive-to-treatment RRMS subjects inversely correlated with disease score and progression. At the cellular level, there was an impaired ANXA1 production by CD4+CD25- conventional T and CD4+RORγt+ T (Th17) cells from RRMS subjects that associated with an increased migratory capacity in an in vitro model of blood brain barrier. Mechanistically, ANXA1 impaired monocyte maturation secondarily to STAT3 hyperactivation and potently reduced T cell activation, proliferation, and glycolysis. Together, these findings identify impaired disease resolution pathways in RRMS caused by dysregulated ANXA1 expression that could represent new potential therapeutic targets in RRMS.


Subject(s)
Annexin A1/immunology , Gene Expression Regulation/immunology , Lymphocyte Activation , Multiple Sclerosis/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Adult , Cell Proliferation , Female , Glycolysis/immunology , Humans , Inflammation/immunology , Inflammation/pathology , Male , Middle Aged , Monocytes/immunology , Monocytes/pathology , Multiple Sclerosis/pathology , STAT3 Transcription Factor/immunology , Severity of Illness Index , Th1 Cells/pathology , Th17 Cells/pathology
4.
Int J Mol Sci ; 22(20)2021 Oct 13.
Article in English | MEDLINE | ID: mdl-34681678

ABSTRACT

The tumor microenvironment (TME) is a dynamic system where nontumor and cancer cells intercommunicate through soluble factors and extracellular vesicles (EVs). The TME in pancreatic cancer (PC) is critical for its aggressiveness and the annexin A1 (ANXA1) has been identified as one of the oncogenic elements. Previously, we demonstrated that the autocrine/paracrine activities of extracellular ANXA1 depend on its presence in EVs. Here, we show that the complex ANXA1/EVs modulates the macrophage polarization further contributing to cancer progression. The EVs isolated from wild type (WT) and ANXA1 knock-out MIA PaCa-2 cells have been administrated to THP-1 macrophages finding that ANXA1 is crucial for the acquisition of a protumor M2 phenotype. The M2 macrophages activate endothelial cells and fibroblasts to induce angiogenesis and matrix degradation, respectively. We have also found a significantly increased presence of M2 macrophage in mice tumor and liver metastasis sections previously obtained by orthotopic xenografts with WT cells. Taken together, our data interestingly suggest the relevance of ANXA1 as potential diagnostic/prognostic and/or therapeutic PC marker.


Subject(s)
Annexin A1/metabolism , Extracellular Vesicles/metabolism , Macrophages/immunology , Neovascularization, Pathologic , Pancreatic Neoplasms/metabolism , Tumor Microenvironment , Animals , Annexin A1/immunology , Cell Line, Tumor , Endothelial Cells/physiology , Fibroblasts/physiology , Humans , Macrophage Activation , Mice , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/physiopathology
5.
Int J Mol Sci ; 22(11)2021 May 30.
Article in English | MEDLINE | ID: mdl-34070747

ABSTRACT

Colorectal cancer (CRC) is one of the main causes of cancer death in the world. Post-translational modifications (PTMs) have been extensively studied in malignancies due to its relevance in tumor pathogenesis and therapy. This review is focused on the dysregulation of glycosyltransferase expression in CRC and its impact in cell function and in several biological pathways associated with CRC pathogenesis, prognosis and therapeutic approaches. Glycan structures act as interface molecules between cells and their environment and in several cases facilitate molecule function. CRC tissue shows alterations in glycan structures decorating molecules, such as annexin-1, mucins, heat shock protein 90 (Hsp90), ß1 integrin, carcinoembryonic antigen (CEA), epidermal growth factor receptor (EGFR), insulin-like growth factor-binding protein 3 (IGFBP3), transforming growth factor beta (TGF-ß) receptors, Fas (CD95), PD-L1, decorin, sorbin and SH3 domain-containing protein 1 (SORBS1), CD147 and glycosphingolipids. All of these are described as key molecules in oncogenesis and metastasis. Therefore, glycosylation in CRC can affect cell migration, cell-cell adhesion, actin polymerization, mitosis, cell membrane repair, apoptosis, cell differentiation, stemness regulation, intestinal mucosal barrier integrity, immune system regulation, T cell polarization and gut microbiota composition; all such functions are associated with the prognosis and evolution of the disease. According to these findings, multiple strategies have been evaluated to alter oligosaccharide processing and to modify glycoconjugate structures in order to control CRC progression and prevent metastasis. Additionally, immunotherapy approaches have contemplated the use of neo-antigens, generated by altered glycosylation, as targets for tumor-specific T cells or engineered CAR (Chimeric antigen receptors) T cells.


Subject(s)
Colorectal Neoplasms/genetics , Glycosphingolipids/immunology , Glycosyltransferases/genetics , Mucins/genetics , Neoplasm Proteins/genetics , Protein Processing, Post-Translational , Annexin A1/genetics , Annexin A1/immunology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Decorin/genetics , Decorin/immunology , ErbB Receptors/genetics , ErbB Receptors/immunology , Gene Expression Regulation, Neoplastic , Glycosphingolipids/metabolism , Glycosylation , Glycosyltransferases/immunology , Humans , Immunotherapy, Adoptive/methods , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/immunology , Integrin beta1/genetics , Integrin beta1/immunology , Microfilament Proteins/genetics , Microfilament Proteins/immunology , Mucins/immunology , Neoplasm Proteins/immunology , fas Receptor/genetics , fas Receptor/immunology
6.
Clin Exp Immunol ; 199(3): 278-293, 2020 03.
Article in English | MEDLINE | ID: mdl-31724735

ABSTRACT

Systemic lupus erythematosus is a chronic inflammatory disease which involves multiple organs. Self-specific B and T cells play a main role in the pathogenesis of lupus and have been defined as a logical target for selective therapy. The protein annexin A1 (ANX A1) is a modulator of the immune system involving many cell types. An abnormal expression of ANX A1 was found on activated B and T cells during autoimmunity, suggesting its importance as a potential therapeutic target. We hypothesize that it may be possible to down-regulate the activity of autoreactive T and B cells from lupus patients in a humanized immunodeficient mouse model by treating them with an antibody against ANX A1. When cultured in the presence of anti-ANX A1, peripheral blood mononuclear cells (PBMC) from lupus patients showed a decreased number of immunoglobulin (Ig)G anti-dsDNA antibody-secreting plasma cells, decreased T cell proliferation and expression of activation markers and increased B and T cell apoptosis. We employed a humanized model of SLE by transferring PBMCs from lupus patients to immunodeficient non-obese diabetic-severe combined immunodeficient (NOD-SCID) mice. The humanized animals presented autoantibodies, proteinuria and immunoglobulin deposition in the renal glomeruli. Treatment of these NOD-SCID mice with an anti-ANX A1 antibody prevented appearance of anti-DNA antibodies and proteinuria, while the phosphate-buffered saline (PBS)-injected animals had high levels after the transfer. The treatment reduced the levels of autoantibodies to several autoantigens, lupus-associated cytokines and disease symptoms.


Subject(s)
Annexin A1/immunology , Antibodies/immunology , B-Lymphocytes/immunology , Disease Models, Animal , Lupus Erythematosus, Systemic/immunology , T-Lymphocytes/immunology , Animals , Annexin A1/metabolism , Antibodies/pharmacology , Antibodies, Antinuclear/immunology , Antibodies, Antinuclear/metabolism , Apoptosis/drug effects , Apoptosis/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lupus Erythematosus, Systemic/metabolism , Lymphocyte Activation/immunology , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
7.
Scand J Immunol ; 92(3): e12915, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32533866

ABSTRACT

Systemic lupus erythematosus is an autoimmune syndrome characterized by the development of autoantibodies to a wide range of antigens. Together with B cells, respective self-reactive T cells have an important contribution in disease progression as being responsible for inflammatory cytokines secretion, B cell activation and promoting amplification of the autoimmune response. Annexin A1 is expressed by many cell types and binds to phospholipids in a Ca2+ -dependent manner. Abnormal expression of annexin A1 was found on activated B and T cells in both murine and human autoimmunity suggesting its potential role as a therapeutic target. In the present study, we have investigated the possibility to suppress autoimmune manifestation in spontaneous mouse model of lupus using anti-annexin A1 antibody. Groups of lupus-prone MRL/lpr mice were treated with the anti-annexin A1 monoclonal antibody, and the disease activity and survival of the animals were following up. Flow cytometry, ELISA assays, and histological and immunofluorescence kidney analyses were used to determine the levels of Annexin A1 expression, cytokines, anti-dsDNA antibodies and kidney injuries. The administration of this monoclonal antibody to MRL/lpr mice resulted in suppression of IgG anti-dsDNA antibody production, modulated IL-10 secretion, decreased disease activity and prolonged survival compared with the control group.


Subject(s)
Annexin A1/antagonists & inhibitors , Annexin A1/immunology , Antibodies, Monoclonal/pharmacology , Immunologic Factors/pharmacology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Animals , Autoantibodies/immunology , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Humans , Immunoglobulin G/immunology , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Inbred MRL lpr , Proteinuria/etiology , Proteinuria/urine , Spleen/immunology , Spleen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Treatment Outcome
8.
Kidney Int ; 96(2): 397-408, 2019 08.
Article in English | MEDLINE | ID: mdl-31142442

ABSTRACT

Granulomatosis with polyangiitis (GPA) is an autoimmune vasculitis associated with anti-neutrophil-cytoplasmic antibodies (ANCA) against proteinase 3 leading to kidney damage. Neutrophils from those patients have increased expression of membrane proteinase 3 during apoptosis. Here we examined whether neutrophils from patients with GPA have dysregulated protein expressions associated with apoptosis. A global proteomic analysis was performed comparing neutrophils from patients with GPA, with healthy individuals under basal conditions and during apoptosis. At disease onset, the cytosolic proteome of neutrophils of patients with GPA before treatment was significantly different from healthy controls, and this dysregulation was more pronounced following ex vivo apoptosis. Proteins involved in cell death/survival were altered in neutrophils of patients with GPA. Several proteins identified were PR3-binding partners involved in the clearance of apoptotic cells, namely calreticulin, annexin-A1 and phospholipid scramblase 1. These proteins form a platform at the membrane of apoptotic neutrophils in patients with GPA but not healthy individuals and this was associated with the clinical presentation of GPA. Thus, our study shows that neutrophils from patients with GPA have an intrinsic dysregulation in proteins involved in apoptotic cell clearance, which could contribute to the unabated inflammation and autoimmunity in GPA. Hence, harnessing these dysregulated pathways could lead to novel biomarkers and targeted therapeutic opportunities to treat kidney disease.


Subject(s)
Annexin A1/metabolism , Apoptosis/immunology , Autoimmunity , Granulomatosis with Polyangiitis/immunology , Neutrophils/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Annexin A1/immunology , Antibodies, Antineutrophil Cytoplasmic/immunology , Biomarkers/metabolism , Calreticulin/immunology , Calreticulin/metabolism , Female , Granulomatosis with Polyangiitis/blood , Granulomatosis with Polyangiitis/diagnosis , Humans , Male , Middle Aged , Myeloblastin/immunology , Myeloblastin/metabolism , Neutrophils/metabolism , Phospholipid Transfer Proteins/immunology , Phospholipid Transfer Proteins/metabolism , Proteomics , Signal Transduction/immunology , Young Adult
9.
J Immunol ; 198(8): 3227-3236, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28289158

ABSTRACT

Leishmaniases are diseases caused by several Leishmania species. Leishmania (Viannia) braziliensis can cause localized cutaneous leishmaniasis (LCL), which heals spontaneously, or mucosal leishmaniasis (ML), characterized by chronic and intense inflammation and scanty parasitism. Annexin A1 (AnxA1) is a protein involved in modulation and resolution of inflammation through multiple mechanisms. In the present study, the role of AnxA1 was investigated in L. braziliensis-infected BALB/c mice. AnxA1 levels increased at the peak of tissue lesion and parasitism in infected mice. AnxA1 increased also after L. braziliensis infection of BALB/c (wild-type [WT]) bone marrow derived macrophages. Despite a lower parasite intake, parasite burden in bone marrow-derived macrophages from AnxA1-/- mice was similar to WT and associated with an early increase of TNF-α and, later, of IL-10. AnxA1-/- mice controlled tissue parasitism similarly to WT animals, but they developed significantly larger lesions at later stages of infection, with a more pronounced inflammatory infiltrate and increased specific production of IFN-γ, IL-4, and IL-10. AnxA1-/- mice also presented higher phosphorylation levels of ERK-1/2 and p65/RelA (NF-κB) and inducible NO synthase expression, suggesting that AnxA1 may be involved in modulation of inflammation in this model of experimental leishmaniasis. Finally, assessment of AnxA1 levels in sera from patients with LCL or ML revealed that ML patients had higher levels of serum AnxA1 than did LCL patients or control subjects. Collectively, these data indicate that AnxA1 is actively expressed during L. braziliensis infection. In the absence of AnxA1, mice are fully able to control parasite replication, but they present more intense inflammatory responses and delayed ability to resolve their lesion size.


Subject(s)
Annexin A1/immunology , Leishmaniasis/immunology , Macrophages/immunology , Adolescent , Adult , Animals , Blotting, Western , Child , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Humans , Inflammation/immunology , Leishmania braziliensis , Leishmaniasis/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Young Adult
10.
Microb Pathog ; 118: 277-284, 2018 May.
Article in English | MEDLINE | ID: mdl-29605649

ABSTRACT

Mast cells (MCs) have important immunoregulatory roles in skin inflammation. Annexin A1 (ANXA1) is an endogenous anti-inflammatory protein that can be expressed by mast cells, neutrophils, eosinophils, monocytes, epithelial and T cells. This study investigated MCs heterogeneity and ANXA1 expression in human dermatoses with special emphasis in leprosy. Sixty one skin biopsies from 2 groups were investigated: 40 newly diagnosed untreated leprosy patients (18 reaction-free, 11 type 1 reaction/T1R, 11 type 2 reaction/T2R); 21 patients with other dermatoses. Tryptase/try+ and chymase/chy + phenotypic markers and toluidine blue stained intact/degranulated MC counts/mm2 were evaluated. Try+/chy+ MCs and ANXA1 were identified by streptavidin-biotin-peroxidase immunostaining and density was reported. In leprosy, degranulated MCs outnumbered intact ones regardless of the leprosy form (from tuberculoid/TT to lepromatous/LL), leprosy reactions (reactional/reaction-free) and type of reaction (T1R/T2R). Compared to other dermatoses, leprosy skin lesions showed lower numbers of degranulated and intact MCs. Try+ MCs outnumbered chy+ in leprosy lesions (reaction-free/reactional, particularly in T2R), but not in other dermatoses. Compared to other dermatoses, ANXA1 expression, which is also expressed in mast cells, was higher in the epidermis of leprosy skin lesions, independently of reactional episode. In leprosy, higher MC degranulation and differential expression of try+/chy+ subsets independent of leprosy type and reaction suggest that the Mycobacterium leprae infection itself dictates the inflammatory MCs activation in skin lesions. Higher expression of ANXA1 in leprosy suggests its potential anti-inflammatory role to maintain homeostasis preventing tissue and nerve damage.


Subject(s)
Annexin A1/biosynthesis , Annexin A1/immunology , Anti-Inflammatory Agents/immunology , Anti-Inflammatory Agents/metabolism , Leprosy/immunology , Leprosy/metabolism , Mast Cells/metabolism , Adult , Aged , Aged, 80 and over , Biopsy , Brazil , Chymases/metabolism , Epidermis/immunology , Epidermis/pathology , Female , Humans , Leprosy/pathology , Leprosy, Lepromatous/metabolism , Leprosy, Tuberculoid/metabolism , Male , Mast Cells/pathology , Middle Aged , Mycobacterium leprae/immunology , Mycobacterium leprae/pathogenicity , Skin/pathology , Skin Diseases/metabolism , Skin Diseases/pathology , Tryptases/metabolism , Young Adult
11.
J Immunol ; 196(4): 1922-32, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26800869

ABSTRACT

Annexin A1 (AnxA1) is a glucocorticoid-regulated protein endowed with anti-inflammatory and proresolving properties. Intact AnxA1 is a 37-kDa protein that may be cleaved in vivo at the N-terminal region by neutrophil proteases including elastase and proteinase-3, generating the 33-kDa isoform that is largely inactive. In this study, we investigated the dynamics of AnxA1 expression and the effects of synthetic (sivelestat [SIV]; Eglin) and natural (secretory leukocyte protease inhibitor [SLPI]; Elafin) protease inhibitors on the resolution of LPS-induced inflammation. During the settings of LPS inflammation AnxA1 cleavage associated closely with the peak of neutrophil and elastase expression and activity. SLPI expression increased during resolving phase of the pleurisy. Therapeutic treatment of LPS-challenge mice with recombinant human SLPI or Elafin accelerated resolution, an effect associated with increased numbers of apoptotic neutrophils in the pleural exudates, inhibition of elastase, and modulation of the survival-controlling proteins NF-κB and Mcl-1. Similar effects were observed with SIV, which dose-dependently inhibited neutrophil elastase and shortened resolution intervals. Mechanistically, SIV-induced resolution was caspase-dependent, associated to increased levels of intact AnxA1 and decreased expression of NF-κB and Mcl-1. The proresolving effect of antiproteases was also observed in a model of monosodium urate crystals-induced inflammation. SIV skewed macrophages toward resolving phenotypes and enhanced efferocytosis of apoptotic neutrophils. A neutralizing antiserum against AnxA1 and a nonselective antagonist of AnxA1 receptor abolished the accelerated resolution promoted by SIV. Collectively, these results show that elastase inhibition not only inhibits inflammation but actually promotes resolution, and this response is mediated by protection of endogenous intact AnxA1 with ensuing augmentation of neutrophil apoptosis.


Subject(s)
Annexin A1/immunology , Inflammation/immunology , Protease Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Flow Cytometry , Glycine/analogs & derivatives , Glycine/pharmacology , Humans , Inflammation/metabolism , Leukocyte Elastase/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neutrophils/immunology , Protease Inhibitors/metabolism , Secretory Leukocyte Peptidase Inhibitor/metabolism , Secretory Leukocyte Peptidase Inhibitor/pharmacology , Sulfonamides/pharmacology
12.
Clin Lab ; 64(6): 895-900, 2018 Jun 01.
Article in English | MEDLINE | ID: mdl-29945332

ABSTRACT

BACKGROUND: Our previous studies revealed that concentrations of circulating antibodies to annexin A1 (ANXA1) were increased in non-small lung cancer (NSCLC). This study was thus designed to replicate this initial finding with an independent sample set. METHODS: An enzyme-linked immunosorbent assay (ELISA) was developed in-house to examine plasma antiANXA1 IgG levels in 220 patients with NSCLC and 200 control subjects. RESULTS: Mann-Whitney U test showed that patients with NSCLC had significantly higher anti-ANXA1 IgG levels than control subjects (Z = -4.02, p < 0.001); male patients appeared to mainly contribute to the increased antibody level (Z = -3.09, p = 0.002). Receiver operating characteristic (ROC) curve analysis showed an overall area under the ROC curve (AUC) of 0.61 (95% CI: 0.56 - 0.67), with sensitivity of 8% against a specificity of 95.0%. Spearman's correlation analysis failed to show a significant correlation between the anti-ANXA1 IgG levels and the expression of three tumor-associated antigens including p53 (r = 0.156, p = 0.027), Ki67 (r = -0.048, p = 0.489), and EGFR (r = 0.02, p = 0.782). CONCLUSIONS: Increased levels of circulating anti-ANXA1 IgG antibody may have a prognostic value for NSCLC.


Subject(s)
Annexin A1/immunology , Antibodies, Anti-Idiotypic/immunology , Biomarkers, Tumor/blood , Carcinoma, Non-Small-Cell Lung/blood , Lung Neoplasms/blood , Aged , Antibodies, Anti-Idiotypic/blood , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/immunology , Female , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/immunology , Male , Middle Aged , Prognosis , ROC Curve
13.
Pharmacol Res ; 119: 278-288, 2017 05.
Article in English | MEDLINE | ID: mdl-28212890

ABSTRACT

Annexin A1 is a multifunctional protein characterised by its actions in modulating the innate and adaptive immune response. Accumulating evidence of altered annexin A1 expression in many human tumours raises interest in its functional role in cancer biology. In breast cancer, altered annexin A1 expression levels suggest a potential influence on tumorigenic and metastatic processes. However, reports of conflicting results reveal a relationship that is much more complex than first conceptualised. In this review, we explore the diverse actions of annexin A1 on breast tumour cells and various host cell types, including stromal immune and structural cells, particularly in the context of cancer immunoediting.


Subject(s)
Annexin A1/immunology , Breast Neoplasms/pathology , Breast/pathology , Adaptive Immunity , Animals , Annexin A1/analysis , Annexin A1/genetics , Annexin A1/metabolism , Breast/immunology , Breast/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Cell Movement , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Immunity, Cellular , Immunity, Innate , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/immunology , Neoplasm Invasiveness/pathology , Tumor Microenvironment
14.
J Immunol ; 194(10): 4940-50, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25876761

ABSTRACT

Glucocorticoid (GC)-induced leucine zipper (GILZ) has been shown to mediate or mimic several actions of GC. This study assessed the role of GILZ in self-resolving and GC-induced resolution of neutrophilic inflammation induced by LPS in mice. GILZ expression was increased during the resolution phase of LPS-induced pleurisy, especially in macrophages with resolving phenotypes. Pretreating LPS-injected mice with trans-activator of transcription peptide (TAT)-GILZ, a cell-permeable GILZ fusion protein, shortened resolution intervals and improved resolution indices. Therapeutic administration of TAT-GILZ induced inflammation resolution, decreased cytokine levels, and promoted caspase-dependent neutrophil apoptosis. TAT-GILZ also modulated the activation of the survival-controlling proteins ERK1/2, NF-κB and Mcl-1. GILZ deficiency was associated with an early increase of annexin A1 (AnxA1) and did not modify the course of neutrophil influx induced by LPS. Dexamethasone treatment resolved inflammation and induced GILZ expression that was dependent on AnxA1. Dexamethasone-induced resolution was not altered in GILZ(-/-) mice due to compensatory expression and action of AnxA1. Our results show that therapeutic administration of GILZ efficiently induces a proapoptotic program that promotes resolution of neutrophilic inflammation induced by LPS. Alternatively, a lack of endogenous GILZ during the resolution of inflammation is compensated by AnxA1 overexpression.


Subject(s)
Inflammation/immunology , Macrophages/immunology , Pleurisy/immunology , Transcription Factors/immunology , Animals , Annexin A1/immunology , Apoptosis/immunology , Blotting, Western , Cell Movement , Disease Models, Animal , Flow Cytometry , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred BALB C , Real-Time Polymerase Chain Reaction
15.
J Immunol ; 195(3): 1139-51, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26101324

ABSTRACT

Blood-derived monocytes remove apoptotic cells and terminate inflammation in settings as diverse as atherosclerosis and Alzheimer's disease. They express high levels of the proresolving receptor ALX/FPR2, which is activated by the protein annexin A1 (ANXA1), found in high abundance in inflammatory exudates. Using primary human blood monocytes from healthy donors, we identified ANXA1 as a potent CD14(+)CD16(-) monocyte chemoattractant, acting via ALX/FPR2. Downstream signaling pathway analysis revealed the p38 MAPK-mediated activation of a calcium independent phospholipase A2 with resultant synthesis of lysophosphatidic acid (LPA) driving chemotaxis through LPA receptor 2 and actin cytoskeletal mobilization. In vivo experiments confirmed ANXA1 as an independent phospholipase A2-dependent monocyte recruiter; congruently, monocyte recruitment was significantly impaired during ongoing zymosan-induced inflammation in AnxA1(-/-) or alx/fpr2/3(-/-) mice. Using a dorsal air-pouch model, passive transfer of apoptotic neutrophils between AnxA1(-/-) and wild-type mice identified effete neutrophils as the primary source of soluble ANXA1 in inflammatory resolution. Together, these data elucidate a novel proresolving network centered on ANXA1 and LPA generation and identify previously unappreciated determinants of ANXA1 and ALX/FPR2 signaling in monocytes.


Subject(s)
Annexin A1/immunology , Apoptosis/immunology , Monocytes/immunology , Neutrophils/immunology , Receptors, Lysophosphatidic Acid/immunology , Actin Cytoskeleton/metabolism , Animals , Annexin A1/genetics , Cells, Cultured , Enzyme Activation/immunology , Humans , Inflammation/immunology , Lipopolysaccharide Receptors/metabolism , Lysophospholipids/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/transplantation , Phospholipases A2, Calcium-Independent/metabolism , RNA Interference , RNA, Small Interfering , Receptors, Formyl Peptide/biosynthesis , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, IgG/metabolism , Receptors, Lysophosphatidic Acid/genetics , Zymosan , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Hepatology ; 60(2): 531-44, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24668763

ABSTRACT

UNLABELLED: Annexin A1 (AnxA1) is an effector of the resolution of inflammation and is highly effective in terminating acute inflammatory responses. However, its role in chronic settings is less investigated. Because changes in AnxA1 expression within adipose tissue characterize obesity in mice and humans, we queried a possible role for AnxA1 in the pathogenesis of nonalcoholic steatohepatitis (NASH), a disease commonly associated with obesity. NASH was induced in wild-type (WT) and AnxA1 knockout (AnxA1 KO) C57BL/6 mice by feeding a methionine-choline deficient (MCD) diet up to 8 weeks. In MCD-fed WT mice, hepatic AnxA1 increased in parallel with progression of liver injury. This mediator was also detected in liver biopsies from patients with NASH and its degree of expression inversely correlated with the extent of fibrosis. In both humans and rodents, AnxA1 production was selectively localized in liver macrophages. NASH in AnxA1 KO mice was characterized by enhanced lobular inflammation resulting from increased macrophage recruitment and exacerbation of the M1 phenotype. Consistently, in vitro addition of recombinant AnxA1 to macrophages isolated from NASH livers down-modulated M1 polarization through stimulation of interleukin-10 production. Furthermore, the degree of hepatic fibrosis was enhanced in MCD-fed AnxA1 KO mice, an effect associated with augmented liver production of the profibrotic lectin, galectin-3. Accordingly, AnxA1 addition to isolated hepatic macrophages reduced galectin-3 expression. CONCLUSIONS: Macrophage-derived AnxA1 plays a functional role in modulating hepatic inflammation and fibrogenesis during NASH progression, suggesting the possible use of AnxA1 analogs for therapeutic control of this disease.


Subject(s)
Annexin A1/immunology , Fatty Liver/immunology , Hepatitis/immunology , Macrophages/immunology , Animals , Annexin A1/genetics , Choline Deficiency/genetics , Choline Deficiency/immunology , Disease Models, Animal , Disease Progression , Fatty Liver/genetics , Hepatitis/genetics , Humans , Liver Cirrhosis/genetics , Liver Cirrhosis/immunology , Male , Methionine/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease , Obesity/genetics , Obesity/immunology
17.
Tumour Biol ; 36(2): 1233-6, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25344217

ABSTRACT

Our recent work demonstrated that circulating levels of IgG antibody to linear peptide antigens derived from annexin A1 (ANXA1) were significantly increased in lung cancer. The present study was then undertaken to test whether circulating anti-ANXA1 antibodies were also altered in breast cancer. An enzyme-linked immunosorbent assay was developed in-house to determine circulating IgG against ANXA1-derived peptide antigens in 152 female patients with breast cancer and 160 female control subjects. Student's t test revealed that patients with breast cancer had significantly higher levels of anti-ANXA1 IgG than control subjects (t = 4.75, P < 0.0001). Receiver operating characteristic (ROC) analysis showed that the area under the ROC curve was 0.73 with 95% confidence interval (CI) 0.67-0.78, and the sensitivity of anti-ANXA1 IgG assay was 23.2% against the specificity of 90%. The levels of anti-ANXA1 IgG did not appear to be stage-dependent, and Pearson correlation analysis showed no correlation between the anti-ANXA1 IgG levels and the stages of breast cancer (r = -0.02, df = 149, P = 0.796). This work suggests that circulating IgG for ANXA1-derived peptide antigens may have both diagnostic and prognostic values for breast cancer although further screening is needed to identify more such peptide antigens derived from tumor-associated antigens.


Subject(s)
Annexin A1/blood , Antibodies/blood , Breast Neoplasms/blood , Neoplastic Cells, Circulating/immunology , Aged , Annexin A1/immunology , Antibodies/immunology , Antibodies, Anti-Idiotypic/blood , Antibodies, Anti-Idiotypic/immunology , Antigens, Neoplasm/blood , Antigens, Neoplasm/immunology , Biomarkers, Tumor/immunology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Female , Humans , Middle Aged , Prognosis
18.
J Immunol ; 190(12): 6478-87, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23686496

ABSTRACT

Endogenous mechanisms regulating the host response during inflammation resolution are critical in ensuring disposal of noxious stimuli and return to homeostasis. In this article, we engineered novel Annexin A1 (AnxA1)-based peptides, AnxA1(2-50), that displayed specific binding to the AnxA1 receptor (formyl peptide receptor 2/Lipoxin A4 receptor [FPR2/ALX]; IC50 ∼4 nM). Intravenous administration of AnxA1(2-50) markedly reduced (>60%) leukocyte adhesion to postcapillary venules in wild type and Fpr1(-/-), but not Fpr2/Alx(-/-), mice. Generation of a metabolically stable form of this peptide (CR-AnxA1(2-50)), engineered by substituting a cleavage site shared by human proteinase 3 and neutrophil elastase, yielded an agonist that was resistant to neutrophil-mediated cleavage and displayed enhanced proresolving actions: accelerated resolution of self-limited inflammation and enhanced macrophage efferocytosis after sterile injury, when compared with AnxA1(2-50). These actions were retained with human primary leukocytes where CR-AnxA1(2-50) decreased neutrophil-endothelial interactions (∼25-45%), and stimulated neutrophil apoptosis and macrophage efferocytosis (∼45%). In murine cardiac ischemia/reperfusion injury, CR-AnxA1(2-50) elicited tissue-protective actions reducing infarct size (∼60%) and incidence of 24-h death. These results identify AnxA1(2-50) and CR-AnxA1(2-50) as FPR2/ALX agonists that harness the proresolving actions of AnxA1, and thus may represent therapeutic tools for treatment of inflammatory conditions.


Subject(s)
Annexin A1/immunology , Anti-Inflammatory Agents/immunology , Inflammation/immunology , Receptors, Formyl Peptide/agonists , Receptors, Formyl Peptide/immunology , Receptors, Lipoxin/agonists , Receptors, Lipoxin/immunology , Animals , Annexin A1/metabolism , Humans , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Knockout , Neutrophils/metabolism , Peptides/immunology , Phagocytosis/immunology
19.
J Immunol ; 190(3): 997-1007, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23267026

ABSTRACT

Annexin A1 (AnxA1) is recognized as an endogenous anti-inflammatory molecule. However, its effects on the adaptive immune response and, in particular, on T cells remain unclear. In this study, we investigated the actions of AnxA1 in three distinct models of T cell-mediated inflammation. In contact hypersensitivity, collagen-induced arthritis, and inflammation induced by OT-II TCR transgenic T cells responding to OVA, AnxA1 deficiency significantly increased Ag-induced T cell proliferation and the resultant level of inflammation. In the contact hypersensitivity model, this was associated with increased adhesion of CD4(+) T cells, CD8(+) T cells, and neutrophils in the dermal microvasculature, as well as increased T cell expression of RORγt and IL-17A. In collagen-induced arthritis, deficiency of endogenous AnxA1 increased susceptibility to arthritis and Ag-specific T cell activation. Deficiency of AnxA1 also increased OVA-induced cutaneous delayed-type hypersensitivity and IFN-γ and IL-17 release. Transfer experiments using CD4(+) T cells from AnxA1(-/-) mice demonstrated that the absence of AnxA1 solely in T cells resulted in increased inflammatory responses in wild-type recipients. Similarly, experiments using AnxA1(-/-) OT-II CD4(+) T cells demonstrated that the absence of AnxA1 in T cells was sufficient to induce increased Ag-specific CD4(+) T cell proliferation in vivo, augment T cell production of IFN-γ, IL-17, TNF, and IL-6, and increase Akt, ERK, and p38 activation. Together, these findings indicate that T cell-expressed AnxA1 functions to attenuate T cell-driven inflammatory responses via T cell-intrinsic effects on intracellular signaling, proliferation, and Th1/Th17 cytokine release.


Subject(s)
Annexin A1/deficiency , CD4-Positive T-Lymphocytes/immunology , Inflammation/immunology , Animals , Annexin A1/immunology , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion , Dermatitis, Allergic Contact/immunology , Dermatitis, Allergic Contact/pathology , Enzyme Activation/immunology , Gene Expression Regulation/immunology , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/pathology , Inflammation/pathology , Interleukin-17/biosynthesis , Interleukin-17/genetics , Lymphocyte Activation , Lymphokines/biosynthesis , Lymphokines/genetics , Lymphokines/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neutrophils/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Ovalbumin/immunology , Ovalbumin/toxicity , Oxazolone/immunology , Oxazolone/toxicity , Peptide Fragments/immunology , Peptide Fragments/toxicity , Signal Transduction/immunology , Specific Pathogen-Free Organisms
20.
J Am Soc Nephrol ; 25(11): 2483-98, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24790181

ABSTRACT

Renal targets of autoimmunity in human lupus nephritis (LN) are unknown. We sought to identify autoantibodies and glomerular target antigens in renal biopsy samples from patients with LN and determine whether the same autoantibodies can be detected in circulation. Glomeruli were microdissected from biopsy samples of 20 patients with LN and characterized by proteomic techniques. Serum samples from large cohorts of patients with systemic lupus erythematosus (SLE) with and without LN and other glomerulonephritides were tested. Glomerular IgGs recognized 11 podocyte antigens, with reactivity varying by LN pathology. Notably, IgG2 autoantibodies against α-enolase and annexin AI were detected in 11 and 10 of the biopsy samples, respectively, and predominated over other autoantibodies. Immunohistochemistry revealed colocalization of α-enolase or annexin AI with IgG2 in glomeruli. High levels of serum anti-α-enolase (>15 mg/L) IgG2 and/or anti-annexin AI (>2.7 mg/L) IgG2 were detected in most patients with LN but not patients with other glomerulonephritides, and they identified two cohorts: patients with high anti-α-enolase/low anti-annexin AI IgG2 and patients with low anti-α-enolase/high anti-annexin AI IgG2. Serum levels of both autoantibodies decreased significantly after 12 months of therapy for LN. Anti-α-enolase IgG2 recognized specific epitopes of α-enolase and did not cross-react with dsDNA. Furthermore, nephritogenic monoclonal IgG2 (clone H147) derived from lupus-prone MRL-lpr/lpr mice recognized human α-enolase, suggesting homology between animal models and human LN. These data show a multiantibody composition in LN, where IgG2 autoantibodies against α-enolase and annexin AI predominate in the glomerulus and can be detected in serum.


Subject(s)
Annexin A1/immunology , Biomarkers, Tumor/immunology , DNA-Binding Proteins/immunology , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Phosphopyruvate Hydratase/immunology , Tumor Suppressor Proteins/immunology , Adolescent , Adult , Animals , Annexin A1/isolation & purification , Autoantibodies/blood , Autoantibodies/immunology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/isolation & purification , Biopsy , DNA-Binding Proteins/genetics , DNA-Binding Proteins/isolation & purification , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Male , Mice, Inbred BALB C , Mice, Inbred MRL lpr , Mice, SCID , Middle Aged , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/isolation & purification , Proteomics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/isolation & purification , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL