Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Ann Oncol ; 34(3): 300-314, 2023 03.
Article in English | MEDLINE | ID: mdl-36494005

ABSTRACT

BACKGROUND: New precision medicine therapies are urgently required for glioblastoma (GBM). However, to date, efforts to subtype patients based on molecular profiles have failed to direct treatment strategies. We hypothesised that interrogation of the GBM tumour microenvironment (TME) and identification of novel TME-specific subtypes could inform new precision immunotherapy treatment strategies. MATERIALS AND METHODS: A refined and validated microenvironment cell population (MCP) counter method was applied to >800 GBM patient tumours (GBM-MCP-counter). Specifically, partition around medoids (PAM) clustering of GBM-MCP-counter scores in the GLIOTRAIN discovery cohort identified three novel patient clusters, uniquely characterised by TME composition, functional orientation markers and immune checkpoint proteins. Validation was carried out in three independent GBM-RNA-seq datasets. Neoantigen, mutational and gene ontology analysis identified mutations and uniquely altered pathways across subtypes. The longitudinal Glioma Longitudinal AnalySiS (GLASS) cohort and three immunotherapy clinical trial cohorts [treatment with neoadjuvant/adjuvant anti-programmed cell death protein 1 (PD-1) or PSVRIPO] were further interrogated to assess subtype alterations between primary and recurrent tumours and to assess the utility of TME classifiers as immunotherapy biomarkers. RESULTS: TMEHigh tumours (30%) displayed elevated lymphocyte, myeloid cell immune checkpoint, programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 transcripts. TMEHigh/mesenchymal+ patients featured tertiary lymphoid structures. TMEMed (46%) tumours were enriched for endothelial cell gene expression profiles and displayed heterogeneous immune populations. TMELow (24%) tumours were manifest as an 'immune-desert' group. TME subtype transitions upon recurrence were identified in the longitudinal GLASS cohort. Assessment of GBM immunotherapy trial datasets revealed that TMEHigh patients receiving neoadjuvant anti-PD-1 had significantly increased overall survival (P = 0.04). Moreover, TMEHigh patients treated with adjuvant anti-PD-1 or oncolytic virus (PVSRIPO) showed a trend towards improved survival. CONCLUSIONS: We have established a novel TME-based classification system for application in intracranial malignancies. TME subtypes represent canonical 'termini a quo' (starting points) to support an improved precision immunotherapy treatment approach.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Glioblastoma/drug therapy , Tumor Microenvironment , Neoplasm Recurrence, Local , Immunotherapy/methods , Brain Neoplasms/drug therapy
2.
Nat Med ; 28(6): 1199-1206, 2022 06.
Article in English | MEDLINE | ID: mdl-35618839

ABSTRACT

Immune checkpoint inhibitors (ICIs) show limited clinical activity in patients with advanced soft-tissue sarcomas (STSs). Retrospective analysis suggests that intratumoral tertiary lymphoid structures (TLSs) are associated with improved outcome in these patients. PEMBROSARC is a multicohort phase 2 study of pembrolizumab combined with low-dose cyclophosphamide in patients with advanced STS (NCT02406781). The primary endpoint was the 6-month non-progression rate (NPR). Secondary endpoints included objective response rate (ORR), progression-free survival (PFS), overall survival (OS) and safety. The 6-month NPR and ORRs for cohorts in this trial enrolling all comers were previously reported; here, we report the results of a cohort enrolling patients selected based on the presence of TLSs (n = 30). The 6-month NPR was 40% (95% confidence interval (CI), 22.7-59.4), so the primary endpoint was met. The ORR was 30% (95% CI, 14.7-49.4). In comparison, the 6-month NPR and ORR were 4.9% (95% CI, 0.6-16.5) and 2.4% (95% CI, 0.1-12.9), respectively, in the all-comer cohorts. The most frequent toxicities were grade 1 or 2 fatigue, nausea, dysthyroidism, diarrhea and anemia. Exploratory analyses revealed that the abundance of intratumoral plasma cells (PCs) was significantly associated with improved outcome. These results suggest that TLS presence in advanced STS is a potential predictive biomarker to improve patients' selection for pembrolizumab treatment.


Subject(s)
Sarcoma , Soft Tissue Neoplasms , Tertiary Lymphoid Structures , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Humans , Retrospective Studies , Sarcoma/drug therapy , Sarcoma/etiology , Soft Tissue Neoplasms/drug therapy , Soft Tissue Neoplasms/etiology , Tertiary Lymphoid Structures/etiology
3.
Eur J Cancer ; 119: 151-157, 2019 09.
Article in English | MEDLINE | ID: mdl-31442817

ABSTRACT

PURPOSE: There are some lines of evidence suggesting a potential role of immunotherapy for treating patients with osteosarcomas. PATIENTS AND METHODS: This was an open-label, multicentre, phase 2 study of pembrolizumab in combination with metronomic cyclophosphamide in patients with advanced osteosarcomas. All patients received 50 mg b.i.d. of cyclophosphamide one week on and one week off and 200 mg of intravenous pembrolizumab (every 3 weeks). There was a dual primary end-point, encompassing both the non-progression and objective responses at 6 months per Response Evaluation Criteria in Solid Tumours (RECIST), version 1.1. An objective response rate of 20% and/or a 6-month non-progression rate of 60% were determined as reasonable objectives for treatment with meaningful effect. Correlative studies of immune biomarkers were planned from the patients' tumour samples. RESULTS: Between October 13 2015 and July 3 2017, 17 patients were included. Fifty were assessable for the efficacy end-point. Four patients experienced tumour shrinkage, resulting in a partial response (PR) in one patient (6.7%). The 6-month non-progression rate was 13.3% (95% confidence interval [CI]: 1.7-40.5). The most frequent adverse events were grade I or II nausea, anaemia, anorexia and fatigue. programmed death-ligand 1 (PD-L1) expression rate was low, observed in only 2 cases of 14 with available tumour material. The only patient who experienced PR had a PD-L1-negative tumour. CONCLUSION: Programmed cell death 1 (PD-1) inhibition has limited activity in osteosarcomas. Further studies investigating PD-1 inhibitor in combination with agents modulating the microenvironment are warranted. TRIAL REGISTRATION: This study is registered with ClinicalTrials.gov, number NCT02406781.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Osteosarcoma/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tumor Microenvironment/drug effects , Administration, Metronomic , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Drug Administration Schedule , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Nausea/chemically induced , Osteosarcoma/metabolism , Osteosarcoma/pathology , Programmed Cell Death 1 Receptor/metabolism , Response Evaluation Criteria in Solid Tumors , Young Adult
4.
Br J Cancer ; 106(12): 1989-96, 2012 Jun 05.
Article in English | MEDLINE | ID: mdl-22669160

ABSTRACT

BACKGROUND: The need to unfold the underlying mechanisms of lung cancer aggressiveness, the deadliest cancer in the world, is of prime importance. Because Fas-associated death domain protein (FADD) is the key adaptor molecule transmitting the apoptotic signal delivered by death receptors, we studied the presence and correlation of intra- and extracellular FADD protein with development and aggressiveness of non-small cell lung cancer (NSCLC). METHODS: Fifty NSCLC patients were enrolled in this prospective study. Intracellular FADD was detected in patients' tissue by immunohistochemistry. Tumours and distant non-tumoural lung biopsies were cultured through trans-well membrane in order to analyse extracellular FADD. Correlation between different clinical/histological parameters with level/localisation of FADD protein has been investigated. RESULTS: Fas-associated death domain protein could be specifically downregulated in tumoural cells and FADD loss correlated with the presence of extracellular FADD. Indeed, human NSCLC released FADD protein, and tumoural samples released significantly more FADD than non-tumoural (NT) tissue (P=0.000003). The release of FADD by both tumoural and NT tissue increased significantly with the cancer stage, and was correlated with both early and late steps of the metastasis process. CONCLUSION: The release of FADD by human NSCLC could be a new marker of poor prognosis as it correlates positively with both tumour progression and aggressiveness.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Fas-Associated Death Domain Protein/metabolism , Lung Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/pathology , Disease Progression , Extracellular Space/metabolism , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis , Prognosis , Prospective Studies
5.
Clin Exp Immunol ; 165(3): 329-37, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21668435

ABSTRACT

A large body of evidence indicates that the immune microenvironment controls tumour development. Primary central nervous system lymphomas (PCNSL) are aggressive tumours growing in the central nervous system (CNS). To evaluate the role and characteristics of this immune-privileged site in anti-tumour defences, we compared the cellular and molecular immune microenvironments of growing murine lymphoma B cells injected into the brain or the spleen. In the brain, immune cells, including dendritic cells and T lymphocytes with a large proportion of CD4(+) forkhead box P3 (FoxP3(+)) regulatory T cells, rapidly infiltrated the tumour microenvironment. These populations also increased in number in the spleen. The T cell cytokine profiles in tumour-bearing mice were similar in the two sites, with predominant T helper type 1 (Th1)/Th17 polarization after polyclonal stimulation, although some interleukin (IL)-4 could also be found. We demonstrated that these T cells have anti-tumour activity in the CNS, although less than in the spleen: nude mice that received lymphoma cells intracerebrally died significantly earlier than immunocompetent animals. These results demonstrate that the brain is able to recruit all the major actors to mount a specific anti-tumour immune response against lymphoma.


Subject(s)
Brain Neoplasms/immunology , Lymphoma, B-Cell/immunology , Splenic Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , Antigen-Presenting Cells/pathology , Brain Neoplasms/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Cell Movement/immunology , Cell Proliferation , Cytokines/metabolism , Dendritic Cells/pathology , Female , Lymphocyte Activation/immunology , Lymphoma, B-Cell/pathology , Mice , Mice, Nude , Splenic Neoplasms/pathology , Survival Analysis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , T-Lymphocytes, Regulatory/pathology , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
6.
Oncogene ; 29(8): 1093-102, 2010 Feb 25.
Article in English | MEDLINE | ID: mdl-19946335

ABSTRACT

The natural history of a tumor includes phases of 'in situ' growth, invasion, extravasation and metastasis. During these phases, tumor cells interact with their microenvironment and are influenced by signals coming from stromal, endothelial, inflammatory and immune cells. Indeed, tumors are often infiltrated by various numbers of lymphocytes, macrophages or mast cells. It is generally believed that the latter produce factors that maintain chronic inflammation and promote tumor growth, whereas lymphocytes may control cancer outcome, as evidenced in mouse models. In this study, we analyze data from large cohorts of human tumors, clearly establishing that infiltration of the primary tumor by memory T cells, particularly of the Th1 and cytotoxic types, is the strongest prognostic factor in terms of freedom from disease and overall survival at all stages of clinical disease. We review data suggesting that tertiary lymphoid structures adjacent to tumors and composed of mature dendritic cells (T and B cells organized as germinal centers) may be the site of an antitumor reaction. We propose an immune scoring based on the type, density and location of lymphocyte infiltrates as a novel prognostic factor for use in addition to tumor node metastasis staging to predict disease-free survival and to aid in decisions regarding adjuvant therapies in early stage human cancers.


Subject(s)
Colorectal Neoplasms/diagnosis , Lymphatic Metastasis/diagnosis , Neoplasm Staging , Prognosis , Animals , Breast Neoplasms/diagnosis , Colorectal Neoplasms/physiopathology , Humans
7.
J Med Genet ; 46(7): 447-50, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19435718

ABSTRACT

BACKGROUND: Deletion of the complement factor H related 1 (CFHR1) gene is a consequence of non-allelic homologous recombination and has been reported to be more frequent in atypical haemolytic uraemic syndrome (aHUS) patients than in the normal population. Therefore, it is considered a susceptibility factor for the disease. aHUS is associated with hereditary or acquired abnormalities that lead to uncontrolled alternative pathway complement activation. We tested the CFHR1 deletion for association with aHUS in a population of French aHUS cases and controls. Furthermore, we examined the effect of the deletion in the context of known aHUS risk factors. METHODS AND RESULTS: 177 aHUS patients and 70 healthy donors were studied. The number of CFHR1 alleles was quantified by multiplex ligation dependant probe amplification (MLPA). The frequency of the deleted allele was significantly higher in aHUS patients than in controls (22.7% vs 8.2%, p<0.001). The highest frequency was in the subgroup of patients exhibiting anti-factor H (FH) autoantibodies (92.9%, p<0.0001 vs controls) and in the group of patients exhibiting a factor I (CFI) gene mutation (31.8%, p<0.001 vs controls). The CFHR1 deletion was not significantly more frequent in the cohort of aHUS patients when patients with anti-FH IgG or CFI mutation were excluded. CONCLUSIONS: The high frequency of CFHR1 deletion in aHUS patients is restricted to the subgroups of patients presenting with anti-FH autoantibodies or, to a lesser degree, CFI mutation. These results suggest that the CFHR1 deletion plays a secondary role in susceptibility to aHUS.


Subject(s)
Complement C3b Inactivator Proteins/genetics , Gene Deletion , Hemolytic-Uremic Syndrome/genetics , Adult , Autoantibodies , Chi-Square Distribution , Child , Cohort Studies , Complement Factor H/immunology , Gene Dosage , Gene Frequency , Genetic Predisposition to Disease , Humans , Mutation , Nucleic Acid Amplification Techniques/methods
8.
Oncogene ; 26(1): 142-7, 2007 Jan 04.
Article in English | MEDLINE | ID: mdl-16799635

ABSTRACT

Tumor necrosis factor receptor (TNFR) associated factor 4 (TRAF4) was initially identified as a gene amplified and overexpressed in breast carcinomas. Our aim was to evaluate whether TRAF4 protein overexpression exists in other cancer types. Immunohistochemistry analysis of tumor samples from 623 patients with 20 different tumor types showed that TRAF4 was overexpressed in 268 tumors (43%), including 82 of 137 lung adenocarcinomas (60%). Interestingly, 32 primary tumors and their matching metastases exhibited mostly similar TRAF4 expression pattern. TRAF4 protein overexpression was limited to cancer cells and the subcellular localization was consistently cytoplasmic in a large majority of cases. To investigate changes in TRAF4 gene copy number, 125 cases from six different types of carcinomas were also analysed by fluorescence in situ hybridization. Out of the 28 cases (22%) showing an increased TRAF4 gene copy number, 23 (82%) were overexpressing the protein. Thus, TRAF4 gene amplification is one of the mechanisms responsible for TRAF4 protein overexpression in human cancers. Considering that TRAF4 is located at 17q11.2 in a region of amplification devoid of known oncogenes and is commonly overexpressed in cancer, our data support an oncogenic role for TRAF4.


Subject(s)
Neoplasms/genetics , TNF Receptor-Associated Factor 4/genetics , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Neoplasms/classification , TNF Receptor-Associated Factor 4/metabolism
9.
J Gen Virol ; 86(Pt 6): 1771-1784, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15914856

ABSTRACT

Measles virus (MV) nucleoprotein (N) is a cytosolic protein that is released into the extracellular compartment after apoptosis and/or secondary necrosis of MV-infected cells in vitro. Thus, MV-N becomes accessible to inhibitory cell-surface receptors: FcgammaRIIB and an uncharacterized nucleoprotein receptor (NR). MV-N is composed of two domains: NCORE (aa 1-400) and NTAIL (aa 401-525). To assess the contribution of MV-N domains and of these two receptors in suppression of cell proliferation, a human melanoma HT144 cell line expressing (HT144IIB1) or lacking FcgammaRIIB1 was used as a model. Specific and exclusive NCORE-FcgammaRIIB1 and NTAIL-NR interactions were shown. Moreover, NTAIL binding to human NR predominantly led to suppression of cell proliferation by arresting cells in the G0/G1 phases of the cell cycle, rather than to apoptosis. NCORE binding to HT144IIB1 cells primarily triggered caspase-3 activation, in contrast to HT144IIB1/IC- cells lacking the FcgammaRIIB1 intra-cytoplasmic tail, thus demonstrating the specific inhibitory effect of the NCORE-FcgammaRIIB1 interaction. MV-N- and NCORE-mediated apoptosis through FcgammaRIIB1 was inhibited by the pan-caspase inhibitor zVAD-FMK, indicating that apoptosis was dependent on caspase activation. By using NTAIL deletion proteins, it was also shown that the region of NTAIL responsible for binding to human NR and for cell growth arrest maps to one of the three conserved boxes (Box1, aa 401-420) found in N of Morbilliviruses. This work unveils novel mechanisms by which distinct domains of MV-N may display different immunosuppressive activities, thus contributing to our comprehension of the immunosuppressive state associated with MV infection. Finally, MV-N domains may be good tools to target tumour cell proliferation and/or apoptosis.


Subject(s)
Antigens, CD/metabolism , Measles virus/physiology , Nucleoproteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, IgG/metabolism , Receptors, Virus/metabolism , Viral Proteins/metabolism , Virus Replication , Animals , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Gene Deletion , Humans , Measles virus/metabolism , Nucleocapsid Proteins , Nucleoproteins/genetics , Protein Structure, Tertiary/genetics , Viral Proteins/genetics
11.
J Immunol ; 167(10): 5814-23, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11698455

ABSTRACT

IgG immune complexes trigger humoral immune responses by cross-linking of FcRs for IgG (FcgammaRs). In the present study, we investigated role of lipid rafts, glycolipid- and cholesterol-rich membrane microdomains, in the FcgammaR-mediated responses. In retinoic acid-differentiated HL-60 cells, cross-linking of FcgammaRs resulted in a marked increase in the tyrosine phosphorylation of FcgammaRIIa, p58(lyn), and p120(c-cbl), which was inhibited by a specific inhibitor of Src family protein tyrosine kinases. After cross-linking, FcgammaRs and tyrosine-phosphorylated proteins including p120(c-cbl) were found in the low-density detergent-resistant membrane (DRM) fractions isolated by sucrose-density gradient ultracentrifugation. The association of FcgammaRs as well as p120(c-cbl) with DRMs did not depend on the tyrosine phosphorylation. When endogenous cholesterol was reduced with methyl-beta-cyclodextrin, the cross-linking did not induce the association of FcgammaRs as well as p120(c-cbl) with DRMs. In addition, although the physical association between FcgammaRIIa and p58(lyn) was not impaired, the cross-linking did not induce the tyrosine phosphorylation. In human neutrophils, superoxide generation induced by opsonized zymosan or chemoattractant fMLP was not affected or increased, respectively, after the methyl-beta-cyclodextrin treatment, but the superoxide generation induced by the insoluble immune complex via FcgammaRII was markedly reduced. Accordingly, we conclude that the cross-linking-dependent association of FcgammaRII to lipid rafts is important for the activation of FcgammaRII-associated Src family protein tyrosine kinases to initiate the tyrosine phosphorylation cascade leading to superoxide generation.


Subject(s)
Membrane Microdomains/metabolism , Membrane Microdomains/physiology , Phosphotyrosine/metabolism , Receptors, IgG/metabolism , Receptors, IgG/physiology , Superoxides/metabolism , Ubiquitin-Protein Ligases , beta-Cyclodextrins , Cells, Cultured , Cyclodextrins/pharmacology , Detergents/chemistry , Enzyme Inhibitors/pharmacology , HL-60 Cells , Humans , Phosphorylation/drug effects , Protein Transport , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-cbl , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Signal Transduction , Tretinoin/pharmacology , src-Family Kinases/metabolism
12.
J Immunol ; 167(9): 5052-60, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11673514

ABSTRACT

Decysin, a gene encoding a disintegrin metalloprotease, is transcribed in human dendritic cells (DC) and germinal centers (GC). We have cloned its murine homologue and show that it is processed by the endoprotease furin before secretion of the catalytic domain. We have defined the cell types that express decysin in mouse spleen in the course of an immune response to T cell-dependent Ags. Like in humans, decysin is transcribed by activated CD11c(+) DC that enter the T cell zone from the marginal zone (MZ). In the GC, decysin is expressed by follicular DC and tingible body macrophages. In addition, a MZ cell population expresses decysin and appears to migrate into the B cell follicle. The majority of these follicle-homing cells express the mannose receptor ligand, a marker for the macrophage-like MZ metallophils. The follicle-homing cells are M-CSF dependent, as they are absent in op/op mice that lack functional M-CSF. This suggests that mannose receptor ligand(+) MZ metallophils differentiate into cells that migrate from the MZ into the B cell follicle. Decysin represents the first marker for this previously unrecognized cell population of the mouse spleen, which may represent a precursor for GCDC and may be specialized in the transport of unprocessed Ag from the MZ into developing GC.


Subject(s)
B-Lymphocytes/metabolism , Lectins, C-Type , Mannose-Binding Lectins , Metalloendopeptidases/biosynthesis , Receptors, Cell Surface/metabolism , ADAM Proteins , Amino Acid Sequence , Animals , Cloning, Molecular , Dendritic Cells/metabolism , Dendritic Cells, Follicular/metabolism , Germinal Center/metabolism , Humans , Ligands , Macrophage Colony-Stimulating Factor/physiology , Macrophages/metabolism , Mannose Receptor , Metalloendopeptidases/genetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Spleen/cytology
13.
J Biol Chem ; 276(19): 16469-77, 2001 May 11.
Article in English | MEDLINE | ID: mdl-11297532

ABSTRACT

Fcgamma receptors mediate antibody-dependent inflammatory responses and cytotoxicity as well as certain autoimmune dysfunctions. Here we report the crystal structure of a human Fc receptor (FcgammaRIIIB) in complex with an Fc fragment of human IgG1 determined from orthorhombic and hexagonal crystal forms at 3.0- and 3.5-A resolution, respectively. The refined structures from the two crystal forms are nearly identical with no significant discrepancies between the coordinates. Regions of the C-terminal domain of FcgammaRIII, including the BC, C'E, FG loops, and the C' beta-strand, bind asymmetrically to the lower hinge region, residues Leu(234)-Pro(238), of both Fc chains creating a 1:1 receptor-ligand stoichiometry. Minor conformational changes are observed in both the receptor and Fc upon complex formation. Hydrophobic residues, hydrogen bonds, and salt bridges are distributed throughout the receptor.Fc interface. Sequence comparisons of the receptor-ligand interface residues suggest a conserved binding mode common to all members of immunoglobulin-like Fc receptors. Structural comparison between FcgammaRIII.Fc and FcepsilonRI.Fc complexes highlights the differences in ligand recognition between the high and low affinity receptors. Although not in direct contact with the receptor, the carbohydrate attached to the conserved glycosylation residue Asn(297) on Fc may stabilize the conformation of the receptor-binding epitope on Fc. An antibody-FcgammaRIII model suggests two possible ligand-induced receptor aggregations.


Subject(s)
Antigens, CD/chemistry , Immunoglobulin Fc Fragments/chemistry , Receptors, IgG/chemistry , Amino Acid Sequence , Antigens, CD/immunology , Binding Sites , Binding Sites, Antibody , Cell Membrane/immunology , Crystallography, X-Ray , GPI-Linked Proteins , Glycosylation , Humans , Immunoglobulin Fc Fragments/immunology , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemistry , Protein Structure, Secondary , Receptors, IgG/immunology , Recombinant Proteins/chemistry , Sequence Alignment , Sequence Homology, Amino Acid
14.
Cancer Res ; 61(5): 2189-93, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11280785

ABSTRACT

Matrix metalloproteinases (MMPs) are extracellular enzymes. Some of them are known to be involved in tumor development and/or progression. Several cellular functions have been proposed for MMPs during malignant processes. Notably, they may be involved in tissue-remodeling processes through their ability to digest matrix components or to participate in tumor neoangiogenesis and, subsequently, in cancer cell proliferation. One of these MMPs, stromelysin-3 (ST3/MMP11), although devoid of enzymatic activity against the matrix components, is associated with human tumor progression and poor patient clinical outcome. Using several in vivo experimental models, it has been demonstrated that ST3 expression by the fibroblastic cells surrounding malignant epithelial cells promotes tumorigenesis in a paracrine manner. The present study was devoted to the identification of the cellular function underlying this ST3-induced tumor promotion using a syngeneic tumorigenesis model in mice. Our results show that ST3 exhibits a new and unexpected role for a MMP, because ST3-increased tumorigenesis does not result from increased neoangiogenesis or cancer cell proliferation but from decreased cancer cell death through apoptosis and necrosis. Thus, during malignancy, the cellular function of ST3 is to favor cancer cell survival in the stromal environment.


Subject(s)
Apoptosis/physiology , Colonic Neoplasms/enzymology , Metalloendopeptidases/deficiency , Animals , Cell Division/physiology , Colonic Neoplasms/blood supply , Colonic Neoplasms/pathology , Inbreeding , Macrophages/immunology , Macrophages/pathology , Matrix Metalloproteinase 11 , Metalloendopeptidases/genetics , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/enzymology , Neutrophils/immunology , Neutrophils/pathology
15.
J Immunol ; 166(5): 3377-83, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11207294

ABSTRACT

We demonstrate that soluble CD16 (sCD16; soluble Fc gamma RIII), a natural ligand of CR3, inhibits the infection of monocytes by primary R5 HIV-1 strain opsonized with serum of seronegative individuals. Inhibition of monocyte infection by sCD16 was similar to that observed with anti-CR3 mAbs, indicating that opsonized HIV may use a CR3-dependent pathway for entry in monocytic cells. Cultured human monocytes express both CR3 (CD11b/CD18) and CCR5 receptors. RANTES, the natural ligand of CCR5, inhibited infection of monocytes with unopsonized HIV particles and partially that of monocytes infected with HIV particles opsonized with complement-derived fragments. Although HIV-infected monocytes from homozygous CCR5 Delta 32/Delta 32 (CCR5(-/-)) individuals produce low levels of p24, cells infected with opsonized particles produced higher levels of p24 than cells infected with unopsonized particles. Our results thus suggest that CR3 may represent an alternative coreceptor to CCR5 of opsonized primary R5 virus entry into monocytes/macrophages. We also observed that the concentration of sCD16 is greatly decreased in sera of HIV-infected patients with low lymphocyte CD4(+) counts. Taken together, our findings suggest that sCD16, present in plasma, may play an important role in controlling HIV-1 spread.


Subject(s)
Antiviral Agents/immunology , CD18 Antigens/physiology , HIV-1/immunology , Macrophage-1 Antigen/physiology , Macrophages/immunology , Monocytes/immunology , Opsonin Proteins/immunology , Receptors, IgG/physiology , Adjuvants, Immunologic/blood , Adjuvants, Immunologic/physiology , Animals , Antiviral Agents/blood , Cell Line , Cells, Cultured , Cricetinae , HIV Infections/blood , HIV Infections/immunology , HIV-1/metabolism , HIV-1/pathogenicity , Humans , Immunosuppressive Agents/blood , Immunosuppressive Agents/immunology , Macrophages/virology , Monocytes/metabolism , Monocytes/virology , Opsonin Proteins/blood , Receptors, CCR5/deficiency , Receptors, CCR5/genetics , Receptors, IgG/blood , Solubility
16.
J Immunol ; 166(1): 531-7, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11123333

ABSTRACT

Immune complex-mediated inflammatory responses are initiated by Fc gamma R on phagocytes. We report in this study that an inhibitory receptor, Fc gamma RIIb2, is expressed on circulating human monocytes, and when co-cross-linked with stimulatory Fc gamma R it down-regulates effector function. Fc gamma RIIb2 expression is increased by IL-4 and decreased by IFN-gamma, in contrast to the activating receptor, Fc gamma RIIa, which is increased by IFN-gamma and decreased by IL-4. Thus, Th1 and Th2 cytokines differentially regulate the opposing Fc gamma R systems, altering the balance of activating and inhibiting Fc gamma R. The detection and cytokine modulation of Fc gamma RIIb2 in human myeloid cells provide evidence of a negative regulator of immune complex-mediated responses in human phagocytes and offer a new approach to limit Ab-triggered inflammation in autoimmune disease.


Subject(s)
Adjuvants, Immunologic/biosynthesis , Cytokines/physiology , Monocytes/immunology , Monocytes/metabolism , Receptors, IgG/biosynthesis , Th1 Cells/immunology , Th2 Cells/immunology , Adjuvants, Immunologic/antagonists & inhibitors , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/physiology , Antigens, CD/genetics , Antigens, CD/metabolism , Blotting, Western , Cells, Cultured , Humans , Interferon-gamma/pharmacology , Interleukin-4/pharmacology , Neutrophils/immunology , Neutrophils/metabolism , Phagocytosis/immunology , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptor Aggregation/immunology , Receptors, IgG/antagonists & inhibitors , Receptors, IgG/genetics , Receptors, IgG/metabolism , Receptors, IgG/physiology , Reverse Transcriptase Polymerase Chain Reaction , Th1 Cells/metabolism , Th2 Cells/metabolism , Transcription, Genetic/immunology , Tumor Cells, Cultured
17.
Immunity ; 13(3): 387-95, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11021536

ABSTRACT

Fc receptors play a major role in immune defenses against pathogens and in inflammatory processes. The crystal structure of a human immunoglobulin receptor, FcgammaRIIIb, has been determined to 1.8 A resolution. The overall fold consists of two immunoglobulin-like domains with an acute interdomain hinge angle of approximately 50 degrees. Trp-113, wedged between the N-terminal D1 and the C-terminal D2 domains, appears to further restrict the hinge angle. The putative Fc binding region of the receptor carries a net positive charge complementary to the negative-charged receptor binding regions on Fc. A 1:1 binding stoichiometry between the receptor and Fc was measured by both the equilibrium and nonequilibrium size-exclusion chromatography. Two separate parallel dimers are observed in the crystal lattice, offering intriguing models for receptor aggregation.


Subject(s)
Extracellular Space/chemistry , Extracellular Space/immunology , Receptors, IgG/chemistry , Amino Acid Sequence , Binding Sites , Chromatography, Gel , Conserved Sequence , Crystallization , Crystallography, X-Ray , Dimerization , Extracellular Space/metabolism , Humans , Immunoglobulin Fragments/chemistry , Ligands , Models, Molecular , Molecular Sequence Data , Oligopeptides/chemistry , Oligopeptides/immunology , Oligopeptides/metabolism , Protein Structure, Tertiary , Receptors, IgG/metabolism , Sequence Homology, Amino Acid
18.
Hum Gene Ther ; 11(12): 1695-703, 2000 Aug 10.
Article in English | MEDLINE | ID: mdl-10954903

ABSTRACT

CD34(+)-derived dendritic cells (DCs) can be infected by the T cell-tropic HIVLAI strain, but are poorly permissive for efficient virus production. However, HIVLAI-infected DCs are able to transmit a vigorous cytopathic infection to activated CD4(+) T cells. We show that DCs differentiated from CD34(+) cells can be efficiently transduced by a retroviral vector carrying the IFN-beta coding sequence. This results in resistance to infection by HIV as shown by a threefold reduction in the HIV DNA copy number per cell, and by inhibition of HIV transmission from DCs to CD4(+) T cells. Moreover, constitutive IFN-beta production by DCs increases the synthesis of IL-12 and IFN-gamma Th1-type cytokines and of the beta-chemokines MIP-1alpha, MIP-1beta, and RANTES. This indicates that IFN-beta transduction of DCs blocks HIV infection and viral transmission to CD4(+) T cells, and could favor cellular immune responses in HIV-infected patients.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Dendritic Cells/virology , Gene Transfer Techniques , HIV Infections/transmission , HIV/pathogenicity , Interferon-beta/genetics , Antigens, CD34/metabolism , Cells, Cultured , Chemokine CCL3 , Chemokine CCL4 , Chemokine CCL5/biosynthesis , Cytopathogenic Effect, Viral , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fetal Blood/cytology , Humans , Interferon-beta/metabolism , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Macrophage Inflammatory Proteins/biosynthesis , Polymerase Chain Reaction
20.
J Mol Biol ; 295(2): 213-24, 2000 Jan 14.
Article in English | MEDLINE | ID: mdl-10623521

ABSTRACT

The binding of multivalent antigen-antibody complexes to receptors for the Fc portion of IgG (FcgammaR) induces the clustering of the FcgammaR and triggers cell activation leading to defence reactions against pathogens. The Fc portion of IgG consists of two identical polypeptide chains which are related to each other by a 2-fold axis and are folded in two structural domains, the C(H)2 domain, near the flexible hinge region of the IgG molecule, and the C(H)3 domain. We studied the interaction in solution between the Fc fragment of mouse IgG2b and the extracellular region of mouse FcgammaRII. We find that one Fc molecule binds one FcgammaRII molecule only. Using NMR spectroscopy, we show that FcgammaRII binds to a negatively charged area of the C(H)2 domain, corresponding to the lower hinge region, and that the binding of FcgammaRII onto one of the two symmetrically related sites on the Fc induces a conformational change in the other site. We therefore propose a model that explains why IgG molecules are unable to trigger FcgammaR-mediated cellular responses spontaneously in the absence of crosslinking by multivalent antigens.


Subject(s)
Receptors, IgG/chemistry , Receptors, IgG/metabolism , Animals , Magnetic Resonance Spectroscopy , Mice , Models, Molecular , Protein Binding , Protein Conformation , Protein Folding , Receptors, IgG/classification , Static Electricity , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...