Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Front Immunol ; 15: 1391404, 2024.
Article in English | MEDLINE | ID: mdl-38799444

ABSTRACT

Introduction: Follicular Lymphoma (FL) results from the malignant transformation of germinal center (GC) B cells. FL B cells display recurrent and diverse genetic alterations, some of them favoring their direct interaction with their cell microenvironment, including follicular helper T cells (Tfh). Although FL-Tfh key role is well-documented, the impact of their regulatory counterpart, the follicular regulatory T cell (Tfr) compartment, is still sparse. Methods: The aim of this study was to characterize FL-Tfr phenotype by cytometry, gene expression profile, FL-Tfr origin by transcriptomic analysis, and functionality by in vitro assays. Results: CD4+CXCR5+CD25hiICOS+ FL-Tfr displayed a regulatory program that is close to classical regulatory T cell (Treg) program, at the transcriptomic and methylome levels. Accordingly, Tfr imprinting stigmata were found on FL-Tfh and FL-B cells, compared to their physiological counterparts. In addition, FL-Tfr co-culture with autologous FL-Tfh or cytotoxic FL-CD8+ T cells inhibited their proliferation in vitro. Finally, although FL-Tfr shared many characteristics with Treg, TCR sequencing analyses demonstrated that part of them derived from precursors shared with FL-Tfh. Discussion: Altogether, these findings uncover the role and origin of a Tfr subset in FL niche and may be useful for lymphomagenesis knowledge and therapeutic management.


Subject(s)
Lymphoma, Follicular , T-Lymphocytes, Regulatory , Lymphoma, Follicular/immunology , Lymphoma, Follicular/genetics , Lymphoma, Follicular/pathology , Humans , T-Lymphocytes, Regulatory/immunology , Gene Expression Profiling , Transcriptome , Tumor Microenvironment/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , T Follicular Helper Cells/immunology , T Follicular Helper Cells/metabolism , Male , Female , Coculture Techniques , Germinal Center/immunology
2.
Front Immunol ; 11: 559866, 2020.
Article in English | MEDLINE | ID: mdl-33133070

ABSTRACT

Fibroblastic reticular cells (FRCs) are the specialized lymphoid stromal cells initially identified as triggering T-cell recruitment and dynamic motion in secondary lymphoid organs. Interestingly, FRCs also display antigen presentation capacities and support lymphocyte survival. CXCR5+CD4+ follicular T cells are important players of B-cell maturation and antibody response. Our study reported that in vitro-differentiated FRC-like cells enhanced the growth of the whole CXCR5+CD4+ T-cell compartment, while enhancing IL-4 secretion specifically by the PD1dimCXCR5+CD4+ cell subset, in a Notch- and ICAM1/LFA1-dependent manner. In addition, we revealed that in follicular lymphoma (FL) tissues, previously identified as enriched for PD1hiCXCR5hiCD4+ mature follicular helper T cells, PD1dimCXCR5+CD4+ T cells displayed an enrichment for Notch and integrin gene signatures, and a Notch and ICAM-1-dependent overexpression of IL-4 compared to their non-malignant counterparts. These findings suggest that the crosstalk between FRCs and CXCR5+PD1dimCD4+ T cells may contribute to the FL IL-4 rich environment, thus providing new insights in FL lymphomagenesis.


Subject(s)
Interleukin-4/biosynthesis , Lymphocyte Activation/immunology , Lymphoid Tissue/cytology , Lymphoid Tissue/physiology , Stromal Cells/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Biomarkers , Cell Communication , Cell Proliferation , Cell Survival , Cytokines/biosynthesis , Gene Expression Profiling , Humans , Immunophenotyping , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Activation/genetics , Receptors, CXCR5/metabolism , Receptors, Notch/metabolism , Transcriptome
3.
Cancers (Basel) ; 12(10)2020 Oct 05.
Article in English | MEDLINE | ID: mdl-33028033

ABSTRACT

Follicular lymphoma (FL), the most frequent indolent non-Hodgkin's B cell lymphoma, is considered as a prototypical centrocyte-derived lymphoma, dependent on a specific microenvironment mimicking the normal germinal center (GC). In agreement, several FL genetic alterations affect the crosstalk between malignant B cells and surrounding cells, including stromal cells and follicular helper T cells (Tfh). In our study, we sought to deconvolute this complex FL supportive synapse by comparing the transcriptomic profiles of GC B cells, Tfh, and stromal cells, isolated from normal versus FL tissues, in order to identify tumor-specific pathways. In particular, we highlighted a high expression of IL-6 and IL-7 in FL B cells that could favor the activation of FL Tfh overexpressing IFNG, able in turn to stimulate FL B cells without triggering MHC (major histocompatibility) class II expression. Moreover, the glycoprotein clusterin was found up-regulated in FL stromal cells and could promote FL B cell adhesion. Finally, besides its expression on Tfh, CD200 was found overexpressed on tumor B cells and could contribute to the induction of the immunosuppressive enzyme indoleamine-2,3 dioxygenase by CD200R-expressing dendritic cells. Altogether our findings led us to outline the contribution of major signals provided by the FL microenvironment and their interactions with malignant FL B cells.

4.
Blood Adv ; 2(15): 1889-1900, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30087107

ABSTRACT

A better characterization of T-cell subsets in the microenvironment of classical Hodgkin lymphoma (cHL) would help to develop immunotherapies. Using multicolor flow cytometry, we identified in 6 of 43 cHL tissue samples a previously unrecognized subset of CD8 T cells coexpressing CXCR5 and inducible T-cell costimulator (ICOS) molecules (CD8CXCR5+ICOS+). These cells shared phenotypic features with follicular helper T (TFH) cells including low CCR7 expression together with high expression of B-cell lymphoma-6, programmed cell death 1, B and T lymphocyte attenuator, CD200, and OX40. They had deficient cytotoxicity, low interferon-γ secretion, and common functional properties with intratumoral CD4+ TFH cells, such as production of interleukin-4 (IL-4), IL-21, CXCL13, and capacity to sustain B cells. Gene profiling analysis showed a significant similarity between the signatures of CD8CXCR5+ICOS+ T cells and CD4+ TFH cells. Benign lymphadenitis tissues (n = 8) were devoid of CD8CXCR5+ICOS+ cells. Among the 35 B-cell lymphoma tissues analyzed, including follicular lymphomas (n = 13), diffuse large cell lymphomas (n = 12), marginal zone lymphomas (MZLs; n = 3), mantle cell lymphomas (n = 3), and chronic lymphocytic leukemias (n = 4), only 1 MZL sample contained CD8CXCR5+ICOS+ cells. Lymphoma tumors with CD8CXCR5+ICOS+ cells shared common histopathological features including residual germinal centers, and contained high amounts of activated CD8CXCR5-ICOS+ cells. These data demonstrate a CD8 T-cell differentiation pathway leading to the acquisition of some TFH similarities. They suggest a particular immunoediting process with global CD8 activation acting mainly, but not exclusively, in HL tumors.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Gene Expression Regulation, Neoplastic , Hodgkin Disease/metabolism , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Neoplasm Proteins/biosynthesis , Receptors, CXCR5/biosynthesis , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation , Cytokines/metabolism , Female , Hodgkin Disease/pathology , Humans , Male
5.
Front Immunol ; 8: 1434, 2017.
Article in English | MEDLINE | ID: mdl-29163507

ABSTRACT

During the last decades, considerable efforts have been done to decipher mechanisms supported by microorganisms or viruses involved in the development, differentiation, and function of immune cells. Pathogens and their associated secretome as well as the continuous inflammation observed in chronic infection are shaping both innate and adaptive immunity. Secondary lymphoid organs are functional structures ensuring the mounting of adaptive immune response against microorganisms and viruses. Inside these organs, germinal centers (GCs) are the specialized sites where mature B-cell differentiation occurs leading to the release of high-affinity immunoglobulin (Ig)-secreting cells. Different steps are critical to complete B-cell differentiation process, including proliferation, somatic hypermutations in Ig variable genes, affinity-based selection, and class switch recombination. All these steps require intense interactions with cognate CD4+ helper T cells belonging to follicular helper lineage. Interestingly, pathogens can disturb this subtle machinery affecting the classical adaptive immune response. In this review, we describe how viruses could act directly on GC B cells, either through B-cell infection or by their contribution to B-cell cancer development and maintenance. In addition, we depict the indirect impact of viruses on B-cell response through infection of GC T cells and stromal cells, leading to immune response modulation.

6.
Blood ; 129(18): 2507-2518, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28202459

ABSTRACT

Follicular lymphoma (FL) is the most frequent indolent lymphoma and is characterized by the accumulation of germinal center-derived malignant B cells engaged in a bidirectional crosstalk with their supportive microenvironment in invaded lymph nodes (LNs) and bone marrow (BM). T follicular helper (TFH) cells and infiltrating stromal cells have been shown to favor FL B-cell growth, but the mechanisms of their protumoral effect and how the LN/BM microenvironment is converted into a lymphoma-permissive cell niche remain poorly understood. We demonstrated here that FL-infiltrating LN and BM stromal cells overexpressed CXCL12 in situ. Interleukin-4 high (IL-4hi) FL-TFH cells, unlike FL B cells themselves, triggered CXCL12 upregulation in human stromal cell precursors. In agreement, expression of CXCL12 was associated with IL-4 expression and signaling within the FL BM and LN niches. This IL-4/CXCL12 axis was amplified in activated lymphoid stromal cells as shown in our in vitro model of human lymphoid stroma differentiation and in an inducible mouse model of ectopic lymphoid organ formation. Finally, CXCL12 triggered primary FL B-cell activation, migration, and adhesion, a process antagonized by BTK and PI3K inhibitors. These data identified the IL-4/CXCL12 loop as a previously unrecognized pathway involved in lymphoid stroma polarization and as a potential therapeutic target in FL patients.


Subject(s)
Bone Marrow/immunology , Chemokine CXCL12/immunology , Interleukin-4/immunology , Lymph Nodes/immunology , Lymphoma, Follicular/immunology , Signal Transduction/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Bone Marrow/pathology , Cell Movement/genetics , Cell Movement/immunology , Chemokine CXCL12/genetics , Female , Humans , Interleukin-4/genetics , Lymph Nodes/pathology , Lymphoma, Follicular/genetics , Lymphoma, Follicular/pathology , Male , Mice , Mice, Knockout , Signal Transduction/genetics , Stromal Cells/immunology , Stromal Cells/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
7.
Clin Biochem ; 49(1-2): 111-6, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26282720

ABSTRACT

BACKGROUND: Histamine (HA) is a small amine playing an important role in anaphylactic reactions. In order to identify and quantify HA in plasma matrix, different methods have been developed but present several disadvantages. Here, we developed an alternative method using liquid chromatography coupled with an ultra-high resolution and accurate mass instrument, Q Exactive™ (Thermo Fisher) (LCHRMS). METHODS: The method includes a protein precipitation of plasma samples spiked with HA-d4 as internal standard (IS). LC separation was performed on a C18 Accucore column (100∗2.1mm, 2.6µm) using a mobile phase containing nonafluoropentanoic acid (3nM) and acetonitrile with 0.1% (v/v) formic acid on gradient mode. Separation of analytes was obtained within 10min. Analysis was performed from full scan mode and targeted MS2 mode using a 5ppm mass window. Ion transitions monitored for targeted MS2 mode were 112.0869>95.0607m/z for HA and 116.1120>99.0855m/z for HA-d4. Calibration curves were obtained by adding standard calibration dilution at 1 to 180nM in TrisBSA. RESULTS: Elution of HA and IS occurred at 4.1min. The method was validated over a range of concentrations from 1nM to 100nM. The intra- and inter-run precisions were <15% for quality controls. Human plasma samples from 30 patients were analyzed by LCHRMS, and the results were highly correlated with those obtained using the gold standard radioimmunoassay (RIA) method. CONCLUSION: Overall, we demonstrate here that LCHRMS is a sensitive method for histamine quantification in biological human plasmas, suitable for routine use in medical laboratories. In addition, LCHRMS is less time-consuming than RIA, avoids the use of radioactivity, and could then be considered as an alternative quantitative method.


Subject(s)
Chromatography, Liquid/methods , Histamine/blood , Mass Spectrometry/methods , Calibration , Humans , Quality Control , Radioimmunoassay , Reference Standards
8.
J Pharm Biomed Anal ; 118: 307-314, 2016 Jan 25.
Article in English | MEDLINE | ID: mdl-26580828

ABSTRACT

Histamine (HA) is one of the main immediate mediators involved in allergic reactions. HA plasma concentration is well correlated with the severity of vascular and respiratory signs of anaphylaxis. Consequently, plasma quantification of HA is useful to comfort the diagnosis of anaphylaxis. Currently, radioimmunoassay (RIA) is the gold standard method to quantify HA due to its high sensitivity, but it is time consuming, implicates specific formations and cautions for technicians, and produces hazardous radioactive wastes. The aim of this study was to compare two enzymatic immunoassays (EIA) and one in-house liquid chromatography high-resolution mass spectrometry method (LC-HRMS) with the gold standard method for HA quantification in plasma samples of patients suspected of anaphylaxis reactions. Ninety-two plasma samples were tested with the 4 methods (RIA, 2 EIA and LC-HRMS) for HA quantification. Fifty-eight samples displayed HA concentrations above the positive cut-off of 10nM evaluated by RIA, including 18 highly positive samples (>100 nM). This study shows that Immunotech(®) EIA and LC-HRMS concentrations were highly correlated with RIA values, in particular for samples with a HA concentration around the positive cut-off. In our hands, plasma concentrations obtained with the Demeditec Diagnostics(®) EIA correlated less with results obtained by RIA, and an underestimation of plasma HA levels led to a lack of sensitivity. In conclusion, this study demonstrates that Immunotech(®) EIA and LC-HRMS method could be used instead of RIA to assess plasma HA in human diagnostic use.


Subject(s)
Histamine/blood , Tandem Mass Spectrometry/methods , Tandem Mass Spectrometry/standards , Chromatography, Liquid/methods , Chromatography, Liquid/standards , Humans , Immunoenzyme Techniques , Mass Spectrometry/methods , Mass Spectrometry/standards , Radioimmunoassay/methods , Radioimmunoassay/standards
9.
Oncotarget ; 6(18): 16471-87, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26158216

ABSTRACT

Both tumor-associated neutrophils (TAN) and cancer-associated fibroblasts (CAFs) display specific phenotypic and functional features and contribute to tumor cell niche. However, their bidirectional crosstalk has been poorly studied, in particular in the context of hematological malignancies. Follicular lymphomas (FL) and diffuse large B-cell lymphomas (DLBCL) are two germinal center-derived lymphomas where various cell components of infiltrating microenvironment, including TAN and CAFs, have been demonstrated to favor directly and indirectly malignant B-cell survival, growth, and drug resistance. We show here that, besides a direct and contact-dependent supportive effect of neutrophils on DLBCL B-cell survival, mediated through the BAFF/APRIL pathway, neutrophils and stromal cells cooperate to sustain FL B-cell growth. This cooperation relies on an overexpression of IL-8 by lymphoma-infiltrating stromal cells that could thereafter efficiently promote neutrophil survival and prime them to neutrophil extracellular trap. Conversely, neutrophils are able to activate stromal cells in a NF-κB-dependent manner, inducing their commitment towards an inflammatory lymphoid stroma phenotype associated with an increased capacity to trigger malignant B-cell survival, and to recruit additional monocytes and neutrophils through the release of CCL2 and IL-8, respectively. Altogether, a better understanding of the lymphoma-supporting effects of neutrophils could be helpful to design new anti-tumor therapeutic strategies.


Subject(s)
B-Lymphocytes/pathology , Fibroblasts/metabolism , Interleukin-8/biosynthesis , Lymphoma, Follicular/pathology , Lymphoma, Large B-Cell, Diffuse/pathology , NF-kappa B/metabolism , Neutrophils/metabolism , Adult , Apoptosis/immunology , B-Cell Activating Factor/antagonists & inhibitors , B-Cell Activating Factor/metabolism , B-Cell Activating Factor/pharmacology , B-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Movement/immunology , Cell Survival/physiology , Chemokine CCL2/metabolism , Child , Extracellular Traps/immunology , Germinal Center , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/metabolism , Interleukin-8/metabolism , Lymphoma, Follicular/immunology , Lymphoma, Large B-Cell, Diffuse/immunology , Neutrophil Infiltration/immunology , Neutrophils/immunology , Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Type I/metabolism , Stromal Cells/metabolism , Tumor Cells, Cultured , Tumor Microenvironment/physiology
10.
Methods Mol Biol ; 1291: 163-73, 2015.
Article in English | MEDLINE | ID: mdl-25836310

ABSTRACT

T-follicular helper (Tfh) cells have emerged as an independent CD4(+) helper T-cell lineage required for antigen-selected germinal center B-cell survival, class switch recombination, and differentiation into long-lived plasma cells. The quantification and function of Tfh subsets are currently extensively explored in humans with infectious diseases, cancer, or autoimmune disorders. Reliable methods to identify and isolate human Tfh cells in patients and healthy donors are necessary to perform these studies. Here, we propose a classical and robust flow cytometric method to detect and isolate Tfh cells from human secondary lymphoid organs based on the expression of CXCR5, PD-1, and CD25 in the CD4(+) T-cell population. An alternative protocol using anti-ICOS and anti-Bcl-6 antibodies and requiring fixation and permeabilization steps without a decrease of detection of membrane markers is also described.


Subject(s)
Cell Separation/methods , Flow Cytometry/methods , Lymph Nodes/cytology , Palatine Tonsil/cytology , T-Lymphocytes, Helper-Inducer/cytology , Humans , Staining and Labeling , Statistics as Topic
11.
Blood ; 125(15): 2381-5, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25733581

ABSTRACT

In follicular lymphoma (FL), follicular helper T cells (TFH) have been depicted as one of the main components of the malignant B-cell niche and a promising therapeutic target. Although defined by their capacity to sustain FL B-cell growth together with specific gene expression and cytokine secretion profiles, FL-TFH constitute a heterogeneous cell population. However, specific markers reflecting such functional heterogeneity are still lacking. In this study, we demonstrate that CD10 identifies a subset of fully functional germinal center TFH in normal secondary lymphoid organs. Importantly, this subset is amplified in the FL context, unlike in other B-cell lymphomas with a follicular growth pattern. Furthermore, whereas FL-TFH produce high levels of interleukin (IL)-21 and low levels of IL-17 irrespectively of their CD10 expression, CD10(pos) FL-TFH specifically exhibit an IL-4(hi)IFN-γ(lo)TNF-α(hi) cytokine profile associated with a high capacity to sustain directly and indirectly malignant B-cell survival. Altogether, our results highlight the important role of this novel functional subset in the FL cell niche.


Subject(s)
B-Lymphocytes/pathology , Germinal Center/cytology , Interleukin-4/immunology , Lymphoma, Follicular/immunology , Neprilysin/immunology , T-Lymphocytes, Helper-Inducer/immunology , B-Lymphocytes/immunology , Cell Survival , Child , Germinal Center/immunology , Germinal Center/pathology , Humans , Interferon-gamma/analysis , Interferon-gamma/immunology , Interleukin-4/analysis , Lymphoma, Follicular/pathology , Neprilysin/analysis , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/immunology
12.
Semin Cancer Biol ; 24: 23-32, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23978491

ABSTRACT

Follicular lymphoma (FL) results from the malignant transformation of germinal center B cells and is characterized by recurrent genetic alterations providing a direct growth advantage or facilitating interaction with tumor microenvironment. In agreement, accumulating evidences suggest a dynamic bidirectional crosstalk between FL B cells and surrounding non-malignant cells within specialized tumor niches in both invaded lymph nodes and bone marrow. Infiltrating stromal cells, macrophages, and T/NK cell subsets either contribute to anti-tumor immune response, or conversely form a tumor supportive network promoting FL B cell survival, growth, and drug resistance. This review depicts the phenotypic heterogeneity and functional plasticity of the most important FL cell partners and describes their complex interplay. We also unravel how malignant B cells recruit and subvert accessory immune and stromal cells to trigger their polarization toward a supportive phenotype. Based on these observations, innovative therapeutic approaches have been recently proposed, in order to benefit from local anti-tumor immunity and/or to selectively target the protective cell niche.


Subject(s)
B-Lymphocytes/pathology , Lymphoma, Follicular/pathology , Tumor Microenvironment/genetics , Cell Communication/genetics , Humans , Lymphoma, Follicular/genetics , Macrophages/pathology , Stromal Cells/pathology , T-Lymphocytes, Helper-Inducer/pathology
13.
PLoS One ; 8(6): e64536, 2013.
Article in English | MEDLINE | ID: mdl-23825522

ABSTRACT

Angioimmunoblastic T-cell Lymphoma (AITL) is one of the most frequent T-cell lymphoma entities. Follicular helper T lymphocytes (TFH) are recognized as the normal cellular counterpart of the neoplastic component. Despite a clonal T-cell feature and few described recurrent cytogenetic abnormalities, a driving oncogenic event has not been identified so far. It has been recently reported that in mice, heterozygous inactivation of Roquin/Rc3h1, a RING type E3 ubiquitine ligase, recapitulates many of the clinical, histological, and cellular features associated with human AITL. In this study we explored whether ROQUIN alterations could be an initial event in the human AITL oncogenic process. Using microarray and RT-PCR analyses, we investigated the levels of ROQUIN transcripts in TFH tumor cells purified from AITL (n = 8) and reactive tonsils (n = 12) and found similar levels of ROQUIN expression in both. Moreover, we also demonstrated that ROQUIN protein was expressed by AITL TFH (PD1+) cells. We then analysed ROQUIN coding sequence in 12 tumor cell-rich AITL samples and found no mutation in any of the samples. Finally, we analysed the expression of MiR101, a putative partner of ROQUIN involved in the modulation of ICOS expression and found similar levels of expression in tumor and reactive TFH. Altogether, this study shows that neither alteration of ROQUIN gene nor deregulation of miR101 expression is likely to be a frequent recurrent event in AITL.


Subject(s)
Immunoblastic Lymphadenopathy/enzymology , Lymphoma, T-Cell/enzymology , Ubiquitin-Protein Ligases/metabolism , Cells, Cultured , Humans , Immunoblastic Lymphadenopathy/genetics , Immunoblastic Lymphadenopathy/metabolism , Immunohistochemistry , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ubiquitin-Protein Ligases/genetics
14.
Front Immunol ; 3: 280, 2012.
Article in English | MEDLINE | ID: mdl-22973275

ABSTRACT

Follicular lymphoma (FL) is the prototypical model of indolent B cell lymphoma displaying a strong dependence on a specialized cell microenvironment mimicking normal germinal center. Within malignant cell niches in invaded lymph nodes and bone marrow, external stimuli provided by infiltrating stromal cells make a pivotal contribution to disease development, progression, and drug resistance. The crosstalk between FL B cells and stromal cells is bidirectional, causing activation of both partners. In agreement, FL stromal cells exhibit specific phenotypic, transcriptomic, and functional properties. This review highlights the critical pathways involved in the direct tumor-promoting activity of stromal cells but also their role in the organization of FL cell niche through the recruitment of accessory immune cells and their polarization to a B cell supportive phenotype. Finally, deciphering the interplay between stromal cells and FL cells provides potential new therapeutic targets with the aim to mobilize malignant cells outside their protective microenvironment and increase their sensitivity to conventional treatment.

15.
Blood ; 119(11): 2556-67, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22289889

ABSTRACT

Accumulating evidence indicates that infiltrating stromal cells contribute directly and indirectly to tumor growth in a wide range of cancers. In follicular lymphoma (FL), malignant B cells are found admixed with heterogeneous lymphoid-like stromal cells within invaded lymph nodes and BM. In addition, mesenchymal stromal cells (MSCs) support in vitro FL B-cell survival, in particular after their engagement toward lymphoid differentiation. We show here that BM-MSCs obtained from patients with FL (FL-MSCs) display a specific gene expression profile compared with MSCs obtained from healthy age-matched donors (HD-MSCs). This FL-MSC signature is significantly enriched for genes associated with a lymphoid-like commitment. Interestingly, CCL2 could be detected at a high level within the FL-cell niche, is up-regulated in HD-MSCs by coculture with malignant B cells, and is overexpressed by FL-MSCs, in agreement with their capacity to recruit monocytes more efficiently than HD-MSCs. Moreover, FL-MSCs and macrophages cooperate to sustain malignant B-cell growth, whereas FL-MSCs drive monocyte differentiation toward a proangiogenic and lipopolysaccharide-unresponsive phenotype close to that of tumor-associated macrophages. Altogether, these results highlight the complex role of FL stromal cells that promote direct tumor B-cell growth and orchestrate FL-cell niche, thus emerging as a potential therapeutic target in this disease.


Subject(s)
B-Lymphocytes/metabolism , Cell Polarity/physiology , Chemokine CCL2/metabolism , Lymphoma, Follicular/pathology , Mesenchymal Stem Cells/cytology , Monocytes/cytology , Stromal Cells/cytology , Adult , Aged , B-Lymphocytes/cytology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , Chemokine CCL2/genetics , Female , Gene Expression Profiling , Humans , Lymphoma, Follicular/etiology , Lymphoma, Follicular/metabolism , Male , Mesenchymal Stem Cells/metabolism , Middle Aged , Monocytes/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Stromal Cells/metabolism
16.
Cancer Res ; 69(7): 3228-37, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19276371

ABSTRACT

Human mesenchymal stem cells (MSC) strongly repress activated T-cell proliferation through the production of a complex set of soluble factors, including the tryptophan-catabolizing enzyme indoleamine 2,3-dioxygenase (IDO), which is induced by IFN-gamma. Conversely, MSCs support survival of follicular lymphoma (FL) B cells, in particular after exposure to tumor necrosis factor-alpha (TNF) and lymphotoxin-alpha1beta2 (LT). The role of MSCs on normal and malignant B-cell growth in steady-state and inflammatory conditions remains to be fully explored. We show here that resting MSCs sustain activated normal B-cell proliferation and survival, whereas IFN-gamma-conditioned MSCs mediate IDO-dependent B-cell growth arrest and apoptosis. IFN-gamma, TNF, and LT are significantly overexpressed by the microenvironment of invaded FL-lymph nodes, but their relative expression patterns are highly heterogeneous between samples. In vitro, IFN-gamma abrogates the B-cell supportive phenotype induced by TNF and LT on MSCs. Moreover, IFN-gamma overrules the growth promoting effect of MSCs on primary purified FL B cells. Altogether, these results underline the crucial role of the cytokine context in the local crosstalk between malignant cells and their microenvironment and provide new insights into our knowledge of the FL cell niche that emerges as a new promising target for innovative therapeutic strategies.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Interferon-gamma/pharmacology , Lymphoma, Follicular/enzymology , Lymphoma, Follicular/pathology , Mesoderm/enzymology , Mesoderm/pathology , B-Lymphocytes/enzymology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Survival/immunology , Child , Humans , Interferon-alpha/immunology , Interferon-alpha/pharmacology , Interferon-gamma/immunology , Lymphocyte Activation , Lymphoma, B-Cell/enzymology , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Lymphoma, Follicular/immunology , Mesoderm/immunology , Stromal Cells/enzymology , Stromal Cells/immunology , Stromal Cells/pathology
17.
J Immunol ; 181(2): 1001-11, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18606651

ABSTRACT

The TNF family member TRAIL is emerging as a promising cytotoxic molecule for antitumor therapy. However, its mechanism of action and the possible modulation of its effect by the microenvironment in follicular lymphomas (FL) remain unknown. We show here that TRAIL is cytotoxic only against FL B cells and not against normal B cells, and that DR4 is the main receptor involved in the initiation of the apoptotic cascade. However, the engagement of CD40 by its ligand, mainly expressed on a specific germinal center CD4(+) T cell subpopulation, counteracts TRAIL-induced apoptosis in FL B cells. CD40 induces a rapid RNA and protein up-regulation of c-FLIP and Bcl-x(L). The induction of these antiapoptotic molecules as well as the inhibition of TRAIL-induced apoptosis by CD40 is partially abolished when NF-kappaB activity is inhibited by a selective inhibitor, BAY 117085. Thus, the antiapoptotic signaling of CD40, which interferes with TRAIL-induced apoptosis in FL B cells, involves NF-kappaB-mediated induction of c-FLIP and Bcl-x(L) which can respectively interfere with caspase 8 activation or mitochondrial-mediated apoptosis. These findings suggest that a cotreatment with TRAIL and an inhibitor of NF-kappaB signaling or a blocking anti-CD40 Ab could be of great interest in FL therapy.


Subject(s)
Apoptosis/immunology , B-Lymphocytes/immunology , CD40 Antigens/immunology , CD40 Ligand/metabolism , Lymphoma, Follicular/immunology , NF-kappa B/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Apoptosis/drug effects , B-Lymphocytes/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/immunology , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD40 Ligand/immunology , Caspase 8/metabolism , Cell Line, Tumor , Cells, Cultured , Humans , Lymphoma, Follicular/metabolism , Lymphoma, Follicular/pathology , NF-kappa B/drug effects , NF-kappa B/immunology , Nitriles/pharmacology , Palatine Tonsil/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Sulfones/pharmacology , Up-Regulation , bcl-X Protein/immunology , bcl-X Protein/metabolism
18.
Blood ; 109(2): 693-702, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-16985173

ABSTRACT

Accumulating evidence indicates that the cellular microenvironment plays a key role in follicular lymphoma (FL) pathogenesis, both within tumor lymph nodes (LNs) and in infiltrated bone marrow where ectopic LN-like reticular cells are integrated within malignant B-cell nodular aggregates. In normal secondary lymphoid organs, specific stromal cell subsets provide a highly specialized microenvironment that supports immune response. In particular, fibroblastic reticular cells (FRCs) mediate immune cell migration, adhesion, and reciprocal interactions. The role of FRCs and their postulated progenitors, that is, bone marrow mesenchymal stem cells (MSCs), in FL remains unexplored. In this study, we investigated the relationships between FRCs and MSCs and their capacity to sustain malignant B-cell growth. Our findings strongly suggest that secondary lymphoid organs contain MSCs able to give rise to adipocytes, chondrocytes, osteoblasts, as well as fully functional B-cell supportive FRCs. In vitro, bone marrow-derived MSCs acquire a complete FRC phenotype in response to a combination of tumor necrosis factor-alpha and lymphotoxin-alpha1beta2. Moreover, MSCs recruit primary FL cells that, in turn, trigger their differentiation into FRCs, making them able to support malignant B-cell survival. Altogether, these new insights into the cross talk between lymphoma cells and their microenvironment could offer original therapeutic strategies.


Subject(s)
Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Lymphoma, Follicular/immunology , Lymphoma, Follicular/pathology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Stromal Cells/physiology , Bone Marrow Cells/cytology , Cell Separation/methods , Cells, Cultured , Humans , Lymphoid Tissue/cytology , Palatine Tonsil/cytology , Stromal Cells/cytology
19.
Exp Hematol ; 33(3): 318-28, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15730855

ABSTRACT

OBJECTIVE: We hypothesized that the presence of tumor cells in bone marrow (BM) could alter hematopoietic progenitor cell functions. Therefore, we evaluated phenotypic and in vitro functional properties of BM-derived CD34+ progenitors issued from untreated and newly diagnosed patients presenting a mature B-lymphoproliferative disorder (LPD) involving the BM (Inv+). PATIENTS AND METHODS: In vitro proliferation and differentiation capacities of primitive and committed progenitors were evaluated by cobblestone area-forming cell (CAFC) and colony-forming cell (CFC) assays, and ex vivo cell expansion. Migratory capacities of CD34+ cells were explored by chemotaxis assays using a CXCL12alpha gradient. RESULTS: Our results showed that CD34+ cells from Inv+ patients overexpressed CD117 and had a significant decrease of week-3 and -6 CAFC, and CFC frequencies, compared to cells obtained from healthy volunteers and LPD patients without BM involvement (Inv-). In addition, progenitors from Inv+ patients maintained a significantly decreased CFC capacity after ex vivo cell expansion, compared to healthy volunteers. However, the former cells held their migratory capacity in response to CXCL12alpha. CONCLUSION: Functional defects of primitive and committed CD34+ progenitors detected among LPD patients with BM tumor involvement suggest either that tumor cells may induced bystander effects on progenitors or that "unusual" CD34+ cells may exist in the BM that could belong to the proliferating tumor tissue.


Subject(s)
Antigens, CD34 , Bone Marrow Neoplasms/physiopathology , Bone Marrow/physiopathology , Bystander Effect , Hematopoietic Stem Cells , Lymphoproliferative Disorders/physiopathology , Adult , Aged , B-Lymphocytes , Bone Marrow Neoplasms/secondary , Case-Control Studies , Female , Humans , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...